Downloaded from orbit.dtu.dk on: Sep 24, 2021

BacHBerry:: BACterial Hosts for production of Bioactive phenolics from bERRY fruits

Dudnik, Alexey; Almeida, A. Filipa; Andrade, Ricardo; Avila, Barbara; Bañados, Pilar; Barbay, Diane; Bassard, Jean Etienne; Benkoulouche, Mounir; Bott, Michael; Braga, Adelaide Total number of authors: 83

Published in: Phytochemistry Reviews

Link to article, DOI: 10.1007/s11101-017-9532-2

Publication date: 2018

Document Version Peer reviewed version

Link back to DTU Orbit

Citation (APA): Dudnik, A., Almeida, A. F., Andrade, R., Avila, B., Bañados, P., Barbay, D., Bassard, J. E., Benkoulouche, M., Bott, M., Braga, A., Breitel, D., Brennan, R., Bulteau, L., Chanforan, C., Costa, I., Costa, R. S., Doostmohammadi, M., Faria, N., Feng, C., ... Forster, J. (2018). BacHBerry:: BACterial Hosts for production of Bioactive phenolics from bERRY fruits. Phytochemistry Reviews, 17(2), 291-326. https://doi.org/10.1007/s11101-017-9532-2

General rights Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights.

 Users may download and print one copy of any publication from the public portal for the purpose of private study or research.  You may not further distribute the material or use it for any profit-making activity or commercial gain  You may freely distribute the URL identifying the publication in the public portal

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

1 TITLE

2 BacHBerry: BACterial Hosts for production of Bioactive phenolics from bERRY fruits

3 RUNNING TITLE

4 BacHBerry

5

6 AUTHORS

7 The list of contributing authors is given on p. 2.

8

9 *Corresponding author:

10 Alexey Dudnik

11 Email: [email protected]

12 Phone: + 45 93 51 11 01

13 Fax: +45 45 25 80 01

14

15

16 Keywords: berries; bioprospecting; microbial cell factories; polyphenols; sustainable production.

17

18 Footnote

19 This article is written by the BacHBerry consortium (www.bachberry.eu) and represents the collective effort 20 of all participating institutions. The authors are therefore listed in alphabetical order.

1

21 The list of authors

22 Alexey Dudnik1,*, A. Filipa Almeida15,20, Ricardo Andrade2, Barbara Avila3, Pilar Bañados3, Diane Barbay4,

23 Jean-Etienne Bassard5, Mounir Benkoulouche4, Michael Bott6, Adelaide Braga7, 21, Dario Breitel12, Rex

24 Brennan10, Laurent Bulteau2, Celine Chanforan8, Inês Costa15,20, Rafael S. Costa9, Mahdi

25 Doostmohammadi9,22, Nuno Faria7, 21, Chengyong Feng17, Armando Fernandes9, Patricia Ferreira7, 21, Roberto

26 Ferro1, Alexandre Foito10, Sabine Freitag10, Gonçalo Garcia15,20, Paula Gaspar1, Joana Godinho-Pereira15,20,

27 Björn Hamberger5, András Hartmann9, Harald Heider4, Carolina Jardim15,20, Alice Julien-Laferriere2, Nicolai

28 Kallscheuer6, Wolfgang Kerbe13, Oscar P. Kuipers11, Shanshan Li17, Nicola Love12, Alberto Marchetti-

29 Spaccamela2, Jan Marienhagen6, Cathie Martin12, Arnaud Mary2, Vincent Mazurek8, Camillo Meinhart13,

30 David Méndez Sevillano14, Regina Menezes15,20, Michael Naesby4, Morten H.H. Nørholm1, Finn T. Okkels8,

31 Joana Oliveira7, 21, Marcel Ottens14, Delphine Parrot2, Lei Pei13, Isabel Rocha7, 21, Rita Rosado-Ramos15,20,

32 Caroline Rousseau4, Marie-France Sagot2, Claudia Nunes dos Santos15,20, Markus Schmidt13, Tatiana

33 Shelenga16, Louise Shepherd10, Ana Rita Silva7, 21, Marcelo Henriques da Silva14, Olivier Simon8, Steen

34 Gustav Stahlhut1, Ana Solopova11, Artem Sorokin16, Derek Stewart10,19, Leen Stougie2, Shang Su17, Vera

35 Thole12, Olga Tikhonova16, Martin Trick12, Philippe Vain12, André Veríssimo9, Ana Vila-Santa9, Susana

36 Vinga9, Michael Vogt6, Liangsheng Wang17, Lijin Wang17, Wei Wei17, Sandra Youssef13, Ana Rute Neves18

37 & Jochen Forster1

38

39 1 The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet,

40 Building 220, 2800 Kgs. Lyngby, Denmark

41 2 Institut National de Recherche en Informatique et Automatique, 43 Boulevard du 11 Novembre 1918,

42 69622 Villeurbanne Cedex, France

43 3 Facultad De Agronomía e Ingeniería Forestal, Pontificia Universidad Católica de Chile, Av. Vicuña

44 Mackenna Ote, 4860 Macul, Chile

45 4 Evolva, Duggingerstrasse 23, 4053 Reinach, Switzerland

46 5 Department of Plant and Environmental Science, University of Copenhagen, 1871 Frederiksberg, Denmark

2

47 6 Forschungszentrum Jülich GmbH, Institut für Bio- und Geowissenschaften, IBG-1: Biotechnologie,

48 Wilhelm-Johnen-Straße, 52428 Jülich, Germany

49 7 Biotempo, Avepark – Edif. Spinpark, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal

50 8 Chr. Hansen Natural Colors A/S, Agern Alle 24, 2970 Hørsholm, Denmark

51 9 IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisboa,

52 Portugal

53 10 The James Hutton Institute, Invergowrie, DD2 5DA Dundee, Scotland, United Kingdom

54 11 Groningen Biomolecular Sciences and Biotechnology Institute, Department of Molecular Genetics,

55 University of Groningen, Linnaeusborg, Nijenborgh 7, 9747 AG Groningen, The Netherlands

56 12 John Innes Centre, Norwich Research Park, NR4 7UH Norwich, United Kingdom

57 13 Biofaction KG, Kundmanngasse 39/12, 1030 Vienna, Austria

58 14 Department of Biotechnology, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, The

59 Netherlands

60 15 Instituto de Biologia Experimental e Tecnológica, Av. República, Qta. do Marquês, 2780-157 Oeiras,

61 Portugal

62 16 Fruit Crops Genetic Resources Department, N. I. Vavilov Research Institute of Plant Industry, B.

63 Morskaya Street 42-44, 190000 St. Petersburg, Russia

64 17 Institute of Botany, The Chinese Academy of Sciences, 20 Nanxincun, Xiangshan, 100093 Beijing, China

65 18 Chr. Hansen Holding A/S, Bøge Alle 10-12, 2970 Hørsholm, Denmark

66 19Institute of Mechanical, Process and Energy Engineering, School of Engineering and Physical Sciences,

67 Heriot Watt University, Edinburgh, Scotland, UK

68 20Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da

69 República, 2780-157 Oeiras, Portugal.

70 21Centre of Biological Engineering, University of Minho, Campus de Gualtar 4710-057, Braga, Portugal

71 22School of Mathematics, University of Edinburgh, EH9 3FD Edinburgh, Scotland, United Kingdom

3

72 Abstract

73 BACterial Hosts for production of Bioactive phenolics from bERRY fruits (BacHBerry) was a three-year

74 project funded by the Seventh Framework Programme (FP7) of the European Union that ran between

75 November 2013 and October 2016. The overall aim of the project was to establish a sustainable and

76 economically-feasible strategy for the production of novel high-value phenolic compounds isolated from

77 berry fruits using bacterial platforms. The project aimed at covering all stages of the discovery and pre-

78 commercialization process, including berry collection, screening and characterization of their bioactive

79 components, identification and functional characterization of the corresponding biosynthetic pathways, and

80 construction of Gram-positive bacterial cell factories producing phenolic compounds. Further activities

81 included optimization of polyphenol extraction methods from bacterial cultures, scale-up of production by

82 fermentation up to pilot scale, as well as societal and economic analyses of the processes. This review article

83 summarizes some of the key findings obtained throughout the duration of the project.

84

4

85 Introduction

86 BacHBerry (BACterial Hosts for production of Bioactive phenolics from bERRY fruits, www.bachberry.eu)

87 has received funding from the European Commission’s Seventh Framework Programme (FP7) under the

88 theme KBBE.2013.3.1-01: “Plant High Value Products - from discovery to final product” with the goal of

89 bringing together experts from the fields of plant biology, industrial biotechnology, analytical chemistry, and

90 social sciences in order to assemble and validate a complete pipeline for discovery and microbial production

91 of novel phenolic compounds of plant origin. The consortium consisted of twelve academic groups, five

92 small and medium sized enterprises and one large enterprise from eleven different countries, namely Austria,

93 Chile, China, Denmark, France, Germany, The Netherlands, Portugal, Russia, Switzerland and the United

94 Kingdom. The diversity of partners has allowed access to a variety of expertise and to plant material that had

95 not been characterized extensively.

96 Polyphenols as a source of bioactive molecules

97 Historically, plants have been not only important sources of nutrients and energy in the human diet but also

98 have provided the fundamentals of traditional and modern medicine. The Plantae kingdom has been

99 estimated to produce more than 200,000 different chemical compounds (Weckwerth 2003), including

100 compounds with proven and potential medical applications. This is reflected in the high number of

101 pharmaceutical products based on, or derived from, plant natural products, such as aspirin, morphine,

102 quinine, paclitaxel and artemisinin (Cragg and Newman 2013). Among the different classes of specialized

103 (i.e. secondary) metabolites phenylpropanoids stand out due to their diversity and ubiquity in the plant

104 kingdom. Fleshy fruits, a rich source of bioactive components, such as ascorbate (vitamin C), terpenoids and

105 polyphenols, are considered to have one the highest antioxidant capacity of commonly eaten foods

106 (Halvorsen et al. 2006). Polyphenol content and composition can vary substantially between fleshy-fruit

107 varieties (Carvalho et al. 2013; Miladinović et al. 2014; Kula et al. 2016), different cultivation conditions

108 (Josuttis et al. 2013; Vagiri et al. 2013; Mazur et al. 2014), harvest times and as a result of different post-

109 harvest factors (Kårlund et al. 2014; Stavang et al. 2015), but particularly between species (Moyer et al.

5

110 2001; McDougall et al. 2008; Ruiz et al. 2010; Marhuenda et al. 2016). Furthermore, plant polyphenols are

111 increasingly being associated with putative bioactivities offering protection against several cardiovascular

112 (Goszcz et al. 2015) and neurological diseases (Figueira et al. in press).

113 The hydroxystilbenes are a group of polyphenols with a C6-C2-C6 skeleton with two or more hydroxyl

114 groups, where a central carbon–carbon double bond is conjugated with two phenolic moieties (Quideau et al.

115 2011; Kasiotis et al. 2013). The most well-known representative of this group is (trans-3,5,4’-

116 hydroxystilbene) (Figure 1a). The compound has gained increasing attention with the discovery of the

117 “French paradox”, an observation that despite a diet with relatively high amounts of saturated fats, French

118 people suffer a relatively low incidence of mortality from coronary heart disease (Renaud and de Lorgeril

119 1992; Catalgol et al. 2012). Like other polyphenols, stilbenes can be further decorated by O-methylation (e.g.

120 resulting in 3-methoxy-resveratrol, or , and 3,5’-dimethoxy-resveratrol, or ),

121 acylation (hydrangenic acid) or by glycosylation (e.g. piceid and ) (Kim et al. 2002; Becker

122 et al. 2003; Zhang et al. 2009; Wang et al. 2015).

123 The flavonoids are one of the largest families of phenolic compounds. Flavonoids are characterized by their

124 basic skeleton composed of three rings (Ghosh and Scheepens 2009), including two benzene rings (A & B)

125 and one heterocyclic ring (C) (Figure 1b). So far, over 5,000 naturally occurring flavonoids have been

126 characterized from various plants and they have been classified into six subgroups, including anthocyanidins

127 (e.g. cyanidin and delphinidin); flavan-3-ols (e.g. catechin and epicatechin); flavonols (e.g. quercetin and

128 kaempferol); flavones (e.g. luteonin and apigenin); isoflavones (e.g. genistein and daidzein), and flavanones

129 (e.g. hesperetin and naringenin).

130 Within each subgroup, considerable variation can exist when it comes to phenolic composition of different

131 fruits, and in particular of berries. Anthocyanin composition provides an excellent example of this diversity:

132 the red-orange color of strawberries is due to the presence of pelargonidin-type anthocyanins in the flesh and

133 skin whereas the deep purple-black color of blackcurrants is due to the accumulation of high amounts of

134 delphinidin and cyanidin-type anthocyanins in the skin.

6

135 Anthocyanidins are flavonoids, which are characterized by a hydroxyl group in position 3 and a C-ring with

136 a positively-charged oxonium ion (Figure 1b). Anthocyanins are water-soluble glycosides of

137 anthocyanidins, in which sugars, consisting of one or more hexoses, are linked to the 3-hydroxyl group of the

138 pyrylium ring (Heldt and Piechulla 2011). These compounds are responsible for the orange/red-to-blue

139 coloration observed in some plants. The most common anthocyanidins include cyanidin (red), delphinidin

140 (bluish-purple), malvinidin (reddish-purple), pelargonidin (orange-red), peonidin (pink) and petunidin

141 (purple). The distribution of anthocyanidins can vary greatly among different berry species (Table 1).

142 Flavonoid compounds frequently are decorated with sugar moieties, sugar-acyl moieties (Giusti and

143 Wrolstad 2003) and can be associated with other flavonoids. The patterns of decoration differ greatly

144 amongst species (see Supplementary Tables S1 and S2).

145 Flavonoids are far more prevalent and diverse in berry species than in other common fruits and vegetables.

146 High flavonol contents are observed in cranberry, lingonberry and blackcurrant (Häkkinen et al. 1999),

147 anthocyanins are the most abundant polyphenol pigments (2-5 g kg-1 fresh weight) in berries (Määttä et al.

148 2001) and many simple phenolic acids are abundant in a wide range of berry species (Herrmann and Nagel

149 1989). Additionally, berries constitute one the most important dietary sources of ellagitannins such as

150 sanguiin H6, lambertianin C (Tö rrö nen 2009; Landete 2011) and condensed tannins such as the

151 proanthocyanidins (Rasmussen et al. 2005; Hellströ m et al. 2009). Stilbenes, in contrast, are not that

152 widespread in berries: resveratrol shows highest abundance in grapes (up to 20-30 mg kg-1 fresh weight), but

153 small amounts of resveratrol, pterostilbene and have been also detected in blueberry, cowberry,

154 cranberry, deerberry and lingonberry (Jeandet et al. 1991; Paredes-López et al. 2010; Manganaris et al.

155 2014). It is clear that soft-fruit species contain a staggering diversity of distinct polyphenol derivatives whose

156 potential is yet to be harnessed.

157 The market for polyphenolic compounds has seen substantial growth over the past few years, and is expected

158 to exceed 850 million USD by 2018 (Aranaz et al. 2016). The major factors contributing to this trend are the

159 growing consumer awareness regarding the benefits of polyphenol consumption and the increasing use of

7

160 polyphenol-containing extracts in food, beverage, and cosmetics products particularly in Asia (Jain and

161 Ramawat 2013; Aranaz et al. 2016; Grand View Research Inc. 2016). The increase in demand also requires

162 efficient and eco-friendly production processes, to improve on current manufacturing practices that mostly

163 rely on extraction from various plant sources (e.g. roots, leaves or fruits) via complex downstream

164 processing (Wang et al. 2016a). The BacHBerry consortium decided to address these challenges and set the

165 following objectives: (i) to systematically analyze the phenolic contents in the large berry germplasm

166 collections available from consortium members, (ii) to establish a publicly-available database of

167 transcriptomic and metabolic data obtained from berry bioprospecting within, as well as outside of the

168 project, (iii) to discover novel bioactivities in berry extracts against a range of human pathologies, such as

169 Alzheimer’s disease and Amyotrophic Lateral Sclerosis (ALS), by high-throughput screening with

170 subsequent identification of functional biomolecules, (iv) to identify the corresponding biosynthetic genes

171 and perform functional characterization of the respective gene products, (v) to assay a selection of the

172 biosynthetic genes for functionality in Gram-positive bacterial hosts and use those to construct bacterial cell

173 factories for phenolic production, (vi) to improve the production efficiency further by introducing

174 modifications to the host metabolic networks predicted via rational design or computational tools developed

175 within the project, (vii) to design and optimize cost-effective food-grade methods for extraction of phenolic

176 compounds from bacterial fermentation broth, and (viii) to optimize fermentation conditions and

177 subsequently upscale the production to pilot plant levels.

178 The project commenced with the selection, sampling and whole-metabolite profiling of berries. In order to

179 explore the potential of the phytochemical diversity present in different berry species, we undertook an

180 untargeted characterization of a wide collection of berries from different species/cultivars utilizing

181 metabolomics-based methods to aid the selection of candidate berry extracts for bioactivity screening.

182 Phytochemical diversity in sampled berries

183 Although significant advances have been made in characterizing the polyphenolic complement of berries,

184 particularly in the context of cultivated species and genotypes, there is limited literature available regarding

8

185 the phytochemical composition of wild and underutilized species/cultivars. Indeed, wild berries are

186 commonly reported as particularly rich in diverse phenolic compounds, often as a result of phenolic-

187 associated astringency having been bred out of cultivated species (Häkkinen et al. 1999). The phenolic

188 diversity of individual berries has been highlighted in many publications (Määttä et al. 2003; Zadernowski et

189 al. 2005; Milivojević et al. 2011), but studies that capture this broad diversity systematically are limited. In

190 BacHBerry, we aimed not only to address this knowledge gap but also to go beyond the state-of-the-art by

191 combining analyses of phenolic diversity with functional testing of berry extracts. The genera targeted in this

192 study included Rubus, Ribes, Vaccinium, Lonicera, Lycium, Aristotelia, Berberis and Ugni collected from

193 different locations in the world including Chile, China, Portugal, Russia and United Kingdom (see

194 Supplementary Table S3).

195 Berry extract metabolite profiling

196 Fruit samples from a total of 112 species/genotypes were collected, extracted, and subsequently subjected to

197 liquid-chromatography mass-spectrometry (LC-MS) metabolomic analysis (see details in Supplementary

198 Materials S1). An untargeted method was used to generate a total of 1,890 mass spectral features (1,506 and

199 384 in positive and negative modes, respectively), which were integrated to generate the dataset used for

200 statistical analysis. Principal component analysis (PCA) was used as a multivariate statistical analysis tool to

201 identify those species, which had the most distinct phytochemical profiles (Figures 2a and 2b).

202 Using the four principal components it was possible to select the species/genotypes, which represented the

203 broadest phenolic diversity (Table 2). In addition to selecting the outlier groups of species, it was also

204 decided to include a small subset of samples (two Rubus idaeus genotypes), which did not separate from the

205 majority of samples in the first principal components. This provided samples with phenolic profiles distinct

206 from the outlier samples and standards for comparison between uncultivated/underutilized species and

207 commonly grown species; in essence a commercial control. The fruits from the selected species (Table 2)

208 were extracted and tested for bioactivity in yeast (Saccharomyces cerevisiae) models for pathological

209 processes associated with several chronic disorders.

9

210 Berry extract bioactivity screening

211 Evidence for the protective role of polyphenols against chronic diseases has increased over the past 20 years

212 (Figueira et al. in press; Goszcz et al. 2015). Neurodegenerative diseases (NDs) represent a group of chronic

213 neurological conditions affecting millions of people worldwide, among which are the Alzheimer's Disease

214 (AD), the Parkinson's Disease (PD), the Huntington's Disease (HD), and Amyotrophic Lateral Sclerosis

215 (ALS). All these diseases have in common the aggregation and deposition of protein aggregates, namely of

216 amyloid β42 (Aβ42) (O’Brien and Wong 2011), αSynuclenin (αSyn) (Shults 2006), huntingtin (HTT)

217 (Miller-Fleming et al. 2008) and FUsed in Sarcoma (FUS) (Kwiatkowski et al. 2009; Vance et al. 2009),

218 which represent the pathological hallmarks of AD, PD, HD and ALS, respectively. In addition, chronic

219 activation of innate immune responses is a process closely associated with neurodegeneration. Its modulation

220 is driven by persistent activation of key transcription factors, such as the Nuclear Factor of Activated T-cells

221 (NFAT) and Nuclear Factor κB (NFκB), which upregulate pro-inflammatory responses creating a positive

222 feedback loop further amplifying initial stimuli. It has been argued that disruption of this loop may represent

223 an important strategy to mitigate the progression of NDs. The pleiotropic effects of polyphenols have been

224 shown to interfere with aggregation-driven neurodegeneration as well as to attenuate chronic inflammatory

225 processes, thereby improving health of cellular and animal models (Figueira et al, in press). Consequently,

226 polyphenol-based therapies represent an underexplored strategy to minimize the huge social and economic

227 impact of NDs.

228 The high degree of evolutionary conservation of fundamental biological processes among eukaryotes has

229 established yeast as a validated model organism to decipher the intricacies of human pathologies, particularly

230 NDs, to identify molecular targets amenable to therapeutic intervention as well as lead molecules with

231 health-promoting potential (Kritzer et al. 2009; Su et al. 2010; Tardiff et al. 2012; Menezes et al. 2015).

232 BacHBerry aimed at identifying phenolic bioactives from harnessing the diversity of phenolics in selected

233 berry germplasm from cultivated, wild and underutilized species of berries. The yeast-based screening

234 platform for bioactivity identification comprised a set of genetically modified S. cerevisiae strains expressing

235 Green Fluorescent Protein (GFP) fused with Aβ42 (Bharadwaj et al. 2010), αSyn (Outeiro and Lindquist

10

236 2003), HTT (Krobitsch and Lindquist 2000) and FUS (Ju et al. 2011) under the control of a galactose-

237 inducible promoter (Figure 3a). Upon activation of expression, these proteins start forming aggregates,

238 which consequently has a negative impact on cellular growth and results in lower GFP fluorescence levels.

239 In case an extract possesses bioactivity against one of the diseases, addition of the extract to the activated

240 yeast cells reduces growth inhibition in the corresponding model. These yeast strains accelerated the

241 identification of phenolic compounds with health-promoting attributes among the most chemically diverse

242 samples identified by the metabolomic analysis (Table 2). For information on the procedures used during the

243 screening see Supplementary Materials S1.

244 An illustrative example of how bioactivities for AD were identified is given in Figure 4a. Upon shift of

245 cells to galactose medium to induce expression of GFP-Aβ42, yeast growth was impaired in comparison to

246 the control strain indicating GFP-Aβ42 proteotoxicity. The treatment with polyphenol-enriched extracts of

247 Lycium chinense significantly recovered growth of these cells revealing its protective role via modulation of

248 Aβ42 toxicity. The bioactivities for PD, HD and ALS were screened using a similar approach, in cells

249 expressing the respective disease proteins.

250 In addition, we have also used a yeast-based model for inflammatory signaling that is based on a Crz1

251 reporter-strain (Prescott et al. 2012; Garcia et al. 2016) (Figure 3b). Crz1 is a yeast orthologue of NFAT,

252 which is an important modulator of inflammation in humans that is known to be involved in development of

253 multiple disorders, such as the inflammatory bowel disease or the rheumatoid arthritis (Pan et al. 2013). Both

254 Crz1 and NFAT are known to be activated by a serine/threonine protein phosphatase calcineurin (CaN) in a

255 Ca2+-dependent manner (Rusnak and Mertz 2000; Bodvard et al. 2013). The utilized reporter strain encodes

256 the β-galactosidase gene (lacZ) under the control of a promoter bearing Crz1-binding sites, the Calcineurin-

257 Dependent Responsive Element (CDRE), allowing the assessment of Crz1 activation through the

258 measurement of β-galactosidase activity using chromogenic substrates (Garcia et al. 2016). Given the

259 evolutionary conservation of NFAT and Crz1 activation mechanisms, in combination with the conserved

260 activity of the immunosuppressant FK506 both in yeast and in humans, the yeast Ca2+/CaN/Crz1 reporter

261 assay represents a reliable tool to identify bioactives with potential to attenuate NFAT-mediated

11

262 inflammatory responses. The potential of Lycium chinense polyphenol-enriched extracts to attenuate

263 inflammation is shown in Figure 4b, exemplifying the approach used in BacHBerry to filter for potential

264 anti-inflammatory activities. The activation of Crz1 by MnCl2, mimicking NFAT activation, led to high β-

265 galactosidase activity and cell treatment with FK506 and Lycium chinense extract strongly reduced the β-

266 galactosidase levels revealing its ability to modulate Crz1, and potentially NFAT, activation.

267 These examples illustrate the strategy used in the BacHBerry project to search for bioactive compounds

268 interfering with pathological processes of NDs and inflammation. The yeast-based screening platform also

269 included strains allowing the identification of metabolites with potential application for type II diabetes,

270 hematological diseases and cancer (unpublished results).

271 Bioassay-guided fractionation compound discovery and candidate compound validation

272 This method of discovering new bioactive natural products ultimately depends on the availability of

273 biological material and preparative- or semi-preparative-scale analytical methods with the capability of

274 resolving mixtures of different classes of compounds typically present in berry extracts (Pauli et al. 2012).

275 Semi-preparative chromatography was used to fractionate selected berry extracts with potential bioactive

276 properties using a hybrid approach of bioassay-guided fractionation procedure (Yang et al. 2001; Weller and

277 G. 2012; Tayarani-Najaran et al. 2013) and screening of pure compounds (Watts et al. 2010). Bioassay-

278 guided fractionation typically involves the following steps: assessment of bioactivity, extraction of the

279 biological material with different solvents and testing of bioactivity. Once bioactivity of an extract has been

280 validated, the extract gets subjected to an iterative process of sub-fractionation/bioactivity testing until pure

281 bioactive natural products are obtained for structural characterization. This approach benefits from exclusion

282 of extracts that do not have bioactivity. However, this procedure requires extensive use of biological material

283 and expensive materials and may result in the isolation of already-known natural products (Duarte et al.

284 2012). Another disadvantage is that the approach is based on the assumption that bioactivity is conferred by

285 a pure compound, although this method can also be used for identification of bioactivities conferred by a

286 cumulative interactions of several polyphenols. Alternatively, pure-compound screening relies on an initial

12

287 isolation and structural elucidation of the individual compounds present in the biological extract followed by

288 bioassay screening. This strategy allows the researcher to focus solely on novel compounds, without re-

289 discovering compounds with well-annotated bioactivity. However, it may also lead to the identification of

290 novel compounds with no bioactivity (Duarte et al. 2012). The second method misses any synergistic

291 interactions affecting bioactivities of berry phenolics.

292 The limited amount of biological material available restricted the number of iterations of fractionation and

293 re-testing of fraction bioactivity possible for typical bioassay-guided fractionation approaches. As described

294 previously, berry extracts typically comprise a relatively diverse and large pool of metabolites that surpass

295 by far the throughput capability of the bioactivity assays used in this study. To overcome these challenges, a

296 hybrid approach was adopted which consisted of several steps: (i) assessment of potential bioactivity present

297 in extracts (as described above), (ii) fractionation of bioactive extracts (see Supplementary Materials S1),

298 (iii) assessment of potential bioactivity present in fractions, (iv) mass-spectrometry-based chemical

299 characterization of bioactive fractions, (v) bioactivity testing of pure candidate compounds (Figure 5).

300 Although this approach shared some of the limitations of the other approaches, it did allow for the exclusion

301 of non-bioactive biological extracts or fractions and focused on the identification novel compounds with

302 potential bioactivities with limited requirement for biological material.

303 Following fractionation of the most promising berry extracts from the first round of screening using the

304 yeast-based platform, isolated fractions were re-analyzed in order to detect bioactive fractions. Subsequently,

305 metabolomic analyses were used for the identification of the individual compounds in each of the bioactive

306 fractions. By utilizing this approach, a set of candidate compounds was defined and re-tested as pure

307 compounds in the yeast-based platform, with further bioactivity validation in the mammalian cell models.

308 Following the BacHBerry pipeline, this information then served as starting point for the engineering of the

309 synthetic pathway for biosynthesis of one of the validated compounds in food-grade bacteria and its

310 subsequent production in small-scale fermenters. The final step of the pipeline, namely re-confirmation of

311 bioactivity of microbially-produced compounds extracted from fermentation broth in both yeast and and

312 mammalian cell models, is currently underway.

13

313 Identification of metabolic pathways and regulators involved in phenolic production

314 The next step within the BacHBerry pipeline involved the identification of the biosynthetic genes and

315 pathways corresponding to the newly discovered bioactives, in order to establish production of these

316 compounds in bacterial hosts. In plants, the biosynthesis of polyphenols occurs via the phenylpropanoid

317 pathway, where the aromatic amino acid L-phenylalanine serves as a precursor. Depending on the enzyme

318 combination, several compounds can be produced in bacteria, including cinnamic acid derivatives, lignin

319 subunits, lignans, phenylpropenes and coumarins, all sharing a basic C6-C3 skeleton (Vogt 2010).

320 Furthermore, the phenylpropanoid backbone can be extended with up to three acetyl-units derived from

321 malonyl-CoA by chalcone synthase (CHS) or stilbene synthase (STS), polyketide synthases which generate

322 various polyketides, such as flavonoids and stilbenes. The core pathways of phenolic biosynthesis are very

323 well understood, with common enzymes, such as, phenylalanine ammonia lyase (PAL), cinnamate 4-

324 hydroxylase (C4H), p-coumaroyl CoA ligase (4CL), hydroxycinnamoyl-CoA shikimate/quinate

325 hydroxycinnamoyltransferase (HCT), p-coumaroyl shikimate/quinate 3'-hydroxylase (C3’H), stilbene

326 synthase and chalcone synthase, found in many plant species (Figure 6).

327 Phenolic chemodiversity in berries, as in other plants, derives from a variety of decorations to the backbone

328 of phenolic structures that include hydroxylations, O-methylations, prenylations, aryl migrations,

329 glycosylations, acylations and polymerizations. For example, anthocyanins are glycosylated on 3-OH

330 positions of the C-ring (Figure 1) and many harbor additional glycosyl groups (for example 5’-O-

331 glycosylation and 3’-O-glycosylation), methoxy groups (petunidin and malvinidin), or hydroxyl groups

332 (which differentiate pelargonidin, cyanidin and delphinidin (Figure 6 and Table 1)). These decorations may

333 affect the bioavailability, bioactivity and stability of polyphenolic compounds. It is therefore essential to

334 develop a greater knowledge of the decorating enzymes specific to berry bioactives, such as those

335 differentially decorating anthocyanins, flavonols, stilbenes, or condensed tannins. Many genes encoding

336 enzymes involved in flavonol and anthocyanin decoration have already been characterized not only in

337 planta, but also heterologously in microorganisms, such as Escherichia coli and S. cerevisiae (Tohge et al.

338 2005; Luo et al. 2007; Pandey et al. 2016; Wang et al. 2016a).

14

339 Berry bioactives identified within the project had special decorations, and those where the genes encoding

340 the decorating activities had not been previously identified, required further research. In a recent work, the

341 transcriptome of the commercial blueberry (Vaccinium corymbosum) was studied using RNA sequencing

342 technology (RNA-seq) in order to discover putative genes related to antioxidant production using next-

343 generation sequencing (NGS) (Li et al. 2012). Similarly, we aimed to identify decorating enzymes and

344 regulators of the biosynthetic pathways of berry bioactives by obtaining transcriptome sequences of target

345 species at different stages of fruit development. This information was used to construct a database of

346 transcriptome sequences for a broad range of berry species, especially those identified as target species

347 containing the promising bioactive molecules. In addition, the overall production of polyphenols is regulated

348 by a range of transcription factors and these were also investigated within the BacHBerry project, with the

349 objective of identifying markers for breeding enhanced levels of phenolic bioactives using the diverse

350 germplasm in selected berry species.

351 Firstly, a library of reference sequences consisting of plant gene sequences encoding enzymes of the core

352 phenolic biosynthetic pathways, which was subsequently used for BLAST searches of berry transcriptomes,

353 was compiled. This library included well-known and experimentally validated anthocyanin, flavonol,

354 stilbene and catechin biosynthetic enzymes from a wide range of plant species. Next, 13 germplasms

355 spanning eight genera, seven families and seven orders were selected for RNA-seq of berry transcriptomes.

356 Sequencing of selected berries at different stages of maturation was also conducted. The data have provided

357 significant resources not only for understanding phenolic biosynthesis in berries, but also for investigating

358 commonalities in genes expressed in berries (Table 3).

359 Multi-layer comparisons of the metabolomics and transcriptomics data of developmental stages, and

360 targeted, as well as, non-targeted searches of candidate transcripts were performed to elucidate genes

361 involved in specific phenolics biosynthetic pathways. Analyses of expression of these genes from different

362 developmental stages of selected berry species were performed with the expVIP tool (Borrill et al. 2016).

15

363 These data were used to search for genes encoding putative decorating enzymes based on the list of

364 molecules of potential interest as bioactives and having special decorations in target berry species.

365 Candidates were identified mostly by searching for homologues of well-characterized plant genes. This was

366 carried out with the help of MassBlast, a software developed within the BacHBerry project

367 (https://github.com/averissimo/mass-blast; (Veríssimo et al. submitted)). We annotated all major enzyme

368 families, including cytochromes P450 (CYP), 2-oxoglutarate-dependent dioxygenases (2-OGDs), UDP-

369 glycosyltransferases (UGTs), BAHD acyltransferases, and O-methyltransferases (OMTs), in all 13 selected

370 berry germplasms. Average counts per transcriptome are presented in Table 4. We also detected candidate

371 transcript(s) for 60-80% of the known biochemical steps of the phenylpropanoid pathway, representing

372 between 109-180 candidates per transcriptome. Approximately 40% of these candidates identified from the

373 transcriptomes were full-length sequences. Furthermore, biochemical activities of some of enzymes encoded

374 by the candidate genes were tested.

375 We also looked for transcripts encoding regulators of phenolic biosynthesis. Since the R2R3-MYB gene

376 family operates extensively in controlling phenylpropanoid metabolism in plants (Stracke et al. 2001;

377 Hartmann et al. 2005), initial searches were focused on identifying MYB-genes that were strongly expressed

378 in berries. However, members of the bHLH and bZIP families of transcription factors were also identified in

379 the berry RNA-seq databases since they may also play roles in regulating accumulation of phenolic

380 bioactives (Hartmann et al. 2005). These data could be particularly useful in breeding programs for selection

381 of plants with improved bioactive contents. The presence of genes encoding putative transporters of phenolic

382 compounds was also examined. Excretion of target compounds produced by bacterial hosts into the culture

383 medium via such transporters would simplify extraction procedures, and may also be necessary in case of

384 any toxic effects of these molecules in bacteria. One transporter is currently being characterized functionally.

385 Gram-positive bacteria as cell factories for the efficient production of berry high-value phenolics

386 While the search for novel bioactives was under way, a set of well-characterized biosynthetic pathway was

387 used for establishing a platform for production of phenolic compounds in Gram-positive bacteria. Two major

16

388 criteria were used for the selection of host organisms for this task: (1) they had to be robust, well-studied

389 organisms with a long-standing history of industrial use and (2) the bacteria must have a generally regarded

390 as safe (GRAS) status and/or be recognized as food-grade. Two industrially-important bacteria, namely

391 Lactococcus lactis and Corynebacterium glutamicum, appeared as attractive candidates for the sustainable

392 production of added-value food and pharmaceutical compounds (Lahtinen et al. 2011; Jojima et al. 2013).

393 Both L. lactis and C. glutamicum are used worldwide in numerous industrial processes, which consequently

394 led to significant amount of research work dedicated towards improving our knowledge of their physiology

395 and genetics. These efforts have resulted in development of large toolboxes for (heterologous) gene

396 expression and introduction of genetic modifications in these bacteria (Burkovski 2008; Pontes et al. 2011;

397 Gaspar et al. 2013). Furthermore, high-throughput methodologies for transcriptome, proteome and

398 metabolome analysis, and genome-scale metabolic models are available for these species (Krömer et al.

399 2004; Oliveira et al. 2005; Shinfuku et al. 2009; Gaspar et al. 2013). Combined with their general robustness,

400 ease of handling and the GRAS status, both L. lactis and C. glutamicum have a strong potential for becoming

401 platform organisms for production of polyphenolic compounds.

402 It should also be noted that during the earlier stages of the project, E. coli was also as a platform for

403 reconstruction of polyphenol biosynthetic pathways. E. coli has a long history of use in research on

404 microbial-based production of various value-added chemicals. Moreover, it is one of the best studied

405 organisms, easy to handle, and there much larger set of genetic tools is available for this organism compared

406 to other bacteria (Yu et al. 2011; Dobson et al. 2012; Chen et al. 2013). Furthermore, E. coli has been

407 previously used for heterologous production of polyphenolic compounds (Pandey et al. 2016; Wang et al.

408 2016a).

409 Lactococcus lactis

410 L. lactis is a Gram-positive bacterium from the order of Lactobacillales that has been widely used in the food

411 industry for the manufacturing of cheese, buttermilk and other dairy products. It is a low G+C aerotolerant

412 bacterium with a relatively small genome and simple sugar metabolism: utilization occurs via glycolysis

413 (Embden–Meyerhof–Parnas pathway) with lactic acid being the major end-product (Gaspar et al. 2013). Its

17

414 ability to ferment various carbohydrates, such as hexoses, pentoses and, in some strains, even complex

415 carbohydrates of plant origin, as well as its high alcohol, acid and stress tolerance makes this bacterium

416 suitable for the harsh environments commonly found under industrial production conditions. L. lactis strains

417 have been engineered to produce multiple commodity chemicals (e.g. 2,3-butanediol and butanol), as well as

418 food additives, including sweeteners (e.g. mannitol and xylitol), flavorings (e.g. acetaldehyde and diacetyl),

419 and vitamins. Furthermore, production of plant secondary metabolites, namely isoprenoids and stilbenes, has

420 been demonstrated in this organism (Neves et al. 2005; Katz et al. 2011; Gaspar et al. 2013; Song et al.

421 2014). In addition, L. lactis is well known as an excellent host for the expression of both prokaryotic and

422 eukaryotic proteins, and has been consequently used for development of live vaccine (Kunji et al. 2005;

423 Hernández et al. 2007; Frelet-Barrand et al. 2010; Pontes et al. 2011).

424 As depicted in Figure 6, in plants the biosynthesis of the core intermediate of the phenylpropanoid pathway,

425 p-coumaric acid, begins from L-phenylalanine and occurs in two steps: a deamination reaction catalyzed by

426 phenylalanine ammonia-lyase (PAL) and a hydroxylation reaction catalyzed by cinnamate 4-hydroxylase

427 (C4H). The latter enzyme belongs to the CYP family that is often challenging to express in active form in

428 prokaryotic systems (Dvora and Koffas 2013). However, in some bacteria and fungi there exists an

429 alternative pathway where L-tyrosine is converted directly into p-coumaric acid via the action of tyrosine

430 ammonia-lyase (TAL), hence bypassing the C4H step (Figure 6). As a first step towards establishing a

431 platform strain for the production of polyphenols in L. lactis, in collaboration with Nielsen and coworkers,

432 we identified several novel TAL enzymes that specifically produce high levels of p-coumaric acid from L-

433 tyrosine (Jendresen et al. 2015).

434 To test the potential of L. lactis to produce more complex phenolic compounds, we introduced a pathway for

435 the biosynthesis of resveratrol. The introduced pathway consisted of three enzymes: TAL, 4CL (4-

436 coumaroyl-CoA ligase, which converts p-coumaric acid to p-coumaroyl-CoA), and STS (stilbene synthase,

437 which catalyzes the condensation of p-coumaroyl-CoA and three molecules of malonyl-CoA to yield trans-

438 resveratrol, Figure 6). The corresponding genes were placed on the high copy number vector pNZ8048

439 under the control of the nisin-inducible promoter PnisA. This promoter is a part of a tightly-controlled

18

440 inducible gene expression system which is based on the auto-regulatory mechanism controlling the

441 production of the bacteriocin nisin (Kuipers et al. 1993; Kuipers et al. 1998). The producer L. lactis strain

442 contains a chromosomally-integrated two-component system NisRK allowing induction of its target

443 promoters by addition of sub-inhibitory concentrations of the bacteriocin nisin to the exponentially-growing

444 cell culture (Linares et al. 2010). Upon cultivation of the producer strain in a chemically defined medium,

445 production of trans-resveratrol was detected in the order of several mg l-1. When TAL was replaced with a

446 promiscuous PAL/TAL enzyme (MacDonald and D’Cunha 2007), additional production of small amounts of

447 (trans-3,5-dihydroxystilbene) from L-phenylalanine was observed. Furthermore, production of

448 methylated variants of trans-resveratrol was also achieved (unpublished results). Obtained titers for trans-

449 resveratrol are comparable to that obtained in an early work on trans-resveratrol production in both E. coli

450 and yeast (Beekwilder et al. 2006; Choi et al. 2011). Current research efforts focus on improving the

451 production efficiency through metabolic engineering of the central carbon metabolism for increased

452 precursor supply.

453 Anthocyanins (glycosylated anthocyanidins) are another group of phenolic phytochemicals. In addition to

454 their antioxidant properties, anthocyanins are valuable for their colors, which range from orange to blue

455 (Falcone Ferreyra et al. 2012). They are of particular interest for the food industry as natural alternatives to

456 replace synthetic dyes. Anthocyanins are synthesized from flavanones by flavanone 3-hydroxylase (F3H),

457 which adds a hydroxyl group to the C3 position. Resulting dihydroflavonols are further processed by

458 dihydroflavonol 4-reductase (DFR), which catalyzes reduction of the carbonyl group of the C4 position to

459 form leucoanthocyanidins, which in turn are converted to anthocyanidins via the action of anthocyanidin

460 synthase (ANS). Anthocyanidins are further stabilized through a C3-glycosylation reaction usually catalyzed

461 by UDP-glucose:flavonoid 3-O-glucosyltransferase (UFGT) to form the anthocyanidin 3-O-glucosides

462 (Falcone Ferreyra et al. 2012) (Figure 6).

463 In order to reconstruct the anthocyanin production pathway in L. lactis, native genes and genes codon-

464 optimized for L. lactis from various plant species coding for F3H, DFR, ANS and UFGT were placed under

465 control of the nisin-inducible promoter PnisA on a high copy-number vector or were integrated into the

19

466 chromosome of L. lactis. Each protein was taggedg with an N- or C- terminal strep-tag, and Western blots

467 were performed to detect the heterologous proteins in cell-free extract of L. lactis. Activity of each enzyme

468 was probed in vitro and in vivo and the reaction products were analyzed using high performance liquid

469 chromatography (HPLC). Production of various types of cyanidin-derived anthocyanidins and anthocyanins

470 by the engineered L. lactis strains was detected, further demonstrating the potential of this lactic acid

471 bacterium as a production host for plant-derived bioactive compounds for food applications (unpublished

472 results). To our knowledge, this is the first report of using lactic acid bacteria for the production of complex

473 phenolic compounds.

474 Malonyl-CoA is a rate-limiting precursor in the biosynthesis of various phenolic compounds in bacteria (Xu

475 et al. 2011; Bhan et al. 2013). Under physiological conditions, L. lactis cells use malonyl-CoA as a substrate

476 in the chain elongation step of the fatty acid biosynthesis. FapR of Bacillus subtilis is a transcriptional

477 repressor that directly senses the intracellular pool of malonyl-CoA. A metabolic biosensor based on the

478 interaction of malonyl-CoA with FapR was previously designed in Escherichia coli (Xu et al. 2013). As a

479 part of the efforts to enhance polyphenol production, a transcriptional biosensor for malonyl-CoA was

480 constructed for the use in C. glutamicum but also proved to be fully functional in L. lactis and could

481 therefore be used for screening of variants with an enhanced malonyl-CoA pool.

482 Among other activities that were pursued in the early stages of the project, we also tried to express plant

483 CYPs in L. lactis, as many steps of the polyphenol biosynthetic pathway are catalyzed by these heme-bound

484 monooxygenases. Functional expression of genes encoding membrane-anchored CYPs in prokaryotes is

485 known to be notoriously difficult, and is limited by problems associated with the differences in intracellular

486 localization (natively to the plant endoplasmatic reticulum), membrane composition, protein folding, and

487 cofactor utilization. The latter element became especially crucial when attempting to express these proteins

488 in L. lactis, because this, normally anaerobic, bacterium does not have the ability to synthetize heme, despite

489 undergoing a metabolic shift to respiration when supplemented with an exogenous source of heme and being

490 capable of regulating heme homeostasis (Pedersen et al. 2012).

20

491 We focused our efforts on CYP79A1 from Sorghum bicolor as it is a well-characterized CYP involved in

492 dhurrin biosynthesis (Bak et al. 2000), and has been previously successfully produced in E. coli (Vazquez-

493 Albacete et al. 2016). As a testbed for the expression of this CYP we have used the Waldo platform (Waldo

494 et al. 1999) that was previously instrumental in producing functional CYP79A1 in E. coli. Briefly, the

495 platform is based on a fusion of the membrane protein with a GFP folding reporter at the C terminus: the

496 fused GFP is designed to act as a fluorescent reporter only when the membrane protein is properly folded

497 and its C terminus is oriented towards the cytoplasm.

498 We detected CYP79A1-GFP by both fluorescence measurements of the L. lactis culture and in-fluorescence

499 protein gels, but were not able to detect activity of the CYP in our in vitro tests. Purified membrane protein

500 fractions from cell cultures grown both aerobically (with hemin supplementation) and anaerobically (with

501 addition of hemin after purification of membrane proteins) did not show any product formation in our TLC-

502 based assay (Blomstedt et al. 2012). These results suggested that CYP79A1 was probably misfolded in L.

503 lactis, although we did not perform any follow-up experiments to confirm this hypothesis. The topic was not

504 pursued any further because any CYP active in the biosynthetic pathways for the selected model compounds

505 could be by-passed (for example the C4H-catalyzed step could be by-passed by using TAL instead of PAL).

506 Corynebacterium glutamicum

507 C. glutamicum is an important organism in industrial biotechnology for the microbial production of bulk

508 chemicals, in particular of amino acids (Becker and Wittmann 2012). C. glutamicum is a very robust

509 microorganism, which shows high resistance to the presence of small aromatic compounds (Liu et al. 2013),

510 which renders C. glutamicum a very promising host for the production of pharmacologically interesting

511 plant-derived polyphenols (Marienhagen and Bott 2013). Until recently, the activity of a complex catabolic

512 network for aromatic compounds meant that C. glutamicum was not used for the production of aromatic

513 compounds (except for aromatic amino acids) (Shen et al. 2012) due to rapid degradation of the products of

514 interest. However, C. glutamicum can be easily modified genetically and a toolbox for the high-level

515 expression of heterologous genes originating from other organisms, has become available, and is based on a

21

516 set of strong and inducible promoters (Eggeling and Bott 2005; Pátek and Nešvera 2013; Kortmann et al.

517 2015).

518 Surprisingly, in initial experiments, it turned out that C. glutamicum is able to grow on naturally occurring

519 phenylpropanoids such as p-coumaric acid, ferulic acid, caffeic acid and 3-(4-hydroxyphenyl)-propionic acid

520 as sole carbon and energy sources. The maximum growth rates on the selected phenylpropanoids ranged

521 from 0.15 to 0.23 h-1 corresponding to doubling times of 3 to 4.5 hours (Kallscheuer et al. 2016a). We

522 suspected that the activity of this pathway would come into conflict with any attempt to produce polyphenols

523 as both pathways compete for phenylpropanoids. Unfortunately, the underlying metabolic pathway

524 responsible for the degradation of phenylpropanoids in C. glutamicum was unknown at the start of the

525 BacHBerry project. Global gene expression analyses were conducted, which led to the identification of a

526 gene cluster consisting of seven genes (designated phdT, phdA, phdR, phdB, phdC, phdD and phdE). In

527 response to feeding of the above-mentioned phenylpropanoids these genes were upregulated up to 100-fold

528 (Kallscheuer et al. 2016a). In subsequent studies, it turned out that the genes in this cluster code for the

529 enzymes of the phenylpropanoid catabolic pathway in C. glutamicum. The gene phdT codes for a

530 phenylpropanoid transporter protein, whereas phdA and the predicted operon phdBCDE code for enzymes

531 involved in the degradation of phenylpropanoids. The phd gene cluster is transcriptionally controlled by a

532 MarR-type repressor encoded by phdR. Cultivation experiments conducted with C. glutamicum strains

533 carrying single-gene deletions showed that loss of phdA, phdB, phdC or phdE abolished growth of C.

534 glutamicum with all tested phenylpropanoid substrates. The intracellular accumulation of pathway

535 intermediates determined via LC-ESI-MS/MS in single-gene deletion mutants showed that the phd gene

536 cluster encodes for enzymes involved in a CoA-dependent, β-oxidative deacetylation pathway, which is

537 essential for the utilization of phenylpropanoids in C. glutamicum. The identified pathway thereby represents

538 a peripheral catabolic route responsible for chain shortening of phenylpropanoids to benzoic acids and

539 acetyl-CoA (Figure 7). C. glutamicum converts the resulting intermediates benzoate, 4-hydroxybenzoate and

540 protocatechuate to succinyl-CoA and acetyl-CoA by central degradation pathways for aromatic compounds

541 (especially the well-characterized β-ketoadipate pathway), and catabolites are ultimately channeled into the

22

542 tricarboxylic acid cycle (Shen and Liu 2005) (Figure 7). Unexpectedly, C. glutamicum failed to grow on the

543 phenylpropanoid, cinnamic acid, which lacks ring para-hydroxylation. Astonishingly, a C. glutamicum strain

544 with a single deletion of the gene phdR coding for the identified transcriptional repressor was able to grow

545 with cinnamic acid as sole carbon and energy source (Kallscheuer et al. 2016a). This indicated that PhdR has

546 a pronounced specificity for hydroxylated phenylpropanoids, especially when hydroxylated in para-position

547 of the aromatic ring. In the course of our studies, cinnamic acid was the only phenylpropanoid tested lacking

548 this hydroxylation, so the complete specificity of this catabolic pathway of phenolics in C. glutamicum

549 remains unknown.

550 During construction of the C. glutamicum platform strain for plant polyphenol production, four gene clusters,

551 comprising 21 genes, including four genes of the phd cluster involved in the catabolism of aromatic

552 compounds, were deleted. The resulting platform strain C. glutamicum DelAro4 was unable to degrade any

553 phenylpropanoid tested, and also could not utilize simple benzoic acids such as protocatechuate (Kallscheuer

554 et al. 2016c). This strain was used as a chassis for the microbial production of polyphenols in particular,

555 stilbenes and (2S)-flavanones, in C. glutamicum.

556 Plasmid-borne expression of codon-optimized genes coding for a 4CL from parsley (Petroselinum crispum),

557 a chalcone synthase (CHS) and a chalcone isomerase (CHI) from Petunia x hybrida from the strong T7

558 promoter in this strain background enabled production of (2S)-flavanones (Figure 7). Maximal titers of 35

559 mg l-1 naringenin and 37 mg l-1 eriodictyol were obtained from the supplemented phenylpropanoids p-

560 coumaric acid and caffeic acid, respectively. Expression of the 4cl gene in combination with a codon-

561 optimized gene coding for an STS from peanut (Arachis hypogaea) allowed for the production of the

562 stilbenes pinosylvin, trans-resveratrol and piceatannol starting from supplemented phenylpropanoids

563 cinnamic acid, p-coumaric acid and caffeic acid, respectively (Kallscheuer et al. 2016c) (Figure 7). Stilbene

564 concentrations of up to 158 mg l-1 could be achieved in defined CGXII medium with 4% glucose and

565 supplemented with cerulenin, a fatty-acid biosynthesis inhibitor that is known to enhance polyphenol

566 production in bacteria by increasing the intracellular concentrations of malonyl-CoA (Santos et al. 2011; van

567 Summeren-Wesenhagen and Marienhagen 2015; Cao et al. 2016). Engineering amino acid metabolism for an

23

568 optimal connection to the synthetic plant polyphenol pathways was conducted to enable stilbene production

569 directly from glucose. Indeed, the additional heterologous expression of aroH from E. coli (coding for the

570 first enzyme of the shikimate pathway responsible for the synthesis of aromatic amino acids) together with a

571 codon-optimized gene coding for a TAL from Flavobacterium johnsoniae led to the production of up to 60

572 mg l-1 trans-resveratrol in the absence of supplemented p-coumaric acid (Kallscheuer et al. 2016c).

573 Furthermore, C. glutamicum was used to produce trans-resveratrol from a cheaper precursor 4-

574 hydroxybenzoic acid (HBA) through reversal of a microbial β-oxidative phenylpropanoid degradation

575 pathway (Kallscheuer et al. 2016b). This novel synthetic pathway circumvents any need for the endogenous

576 supply of aromatic amino acids as polyphenol precursors and in C. glutamicum instantly yielded 5 mg l-1

577 trans-resveratrol from supplemented HBA without any further optimization. Very recently, we also

578 engineered C. glutamicum to produce more complex polyphenols including flavonols (kaempferol and

579 quercetin) and O-methylated stilbenes (pinostilbene and pterostilbene). The obtained pterostilbene titer was

580 42 mg l-1, which is comparable to the titer of 50 mg l-1obtained in engineered E. coli. In C. glutamicum, for

581 the flavonols kaempferol and quercetin titers of 23 mg l-1 and 10 mg l-1, respectively, were detected. The

582 aforementioned titers were obtained from the phenylpropanoids p-coumaric acid and caffeic acid ,

583 respectively, as precursors and represent the highest flavonol titers obtained in engineered microorganism

584 until today (Kallscheuer et al. 2017).

585 The main bottlenecks for the production of polyphenols turned out to be the TAL activity as well as the

586 intracellular availability of malonyl-CoA, representing the cosubstrate in the STS- and CHS-reactions. On-

587 going efforts to improve polyphenol production in C. glutamicum therefore focus mainly on improving the

588 activity of the rate-limiting enzymes and increasing the intracellular malonyl-CoA pool.

589 Other activities

590 As a part of this work, we also aimed at reconstruction of a previously unknown biosynthetic pathway for the

591 flavonol fisetin (3,7,3′,4′-tetrahydroxyflavone, Figure 8). Fisetin is a potent anti-inflammatory, anti-cancer,

592 and anti-oxidant compound (Suh et al. 2008; Funakoshi-Tago et al. 2011; Jash and Mondal 2014). Moreover,

24

593 it has been demonstrated to have a neuroprotective effect and also to limit the complications of type I

594 diabetes (Maher 2009; Maher et al. 2011). Based on the molecular structure of this compound, we

595 hypothesized that its biosynthesis should follow the general pathway of flavonol biosynthesis (Figures 6 and

596 8): a flavanone precursor would be converted into dihydroflavonol by F3H, and then further converted into

597 the corresponding flavonol via the action of flavonol synthase (FLS). In the last step of the biosynthesis, a

598 hydroxyl group would be added at the C3’ position by flavonoid 3’-hydroxylase (F3’H). This part of the

599 work was carried out in E. coli. Isoliquiritigenin (2',4',4-trihydroxychalcone) appeared to be the most suitable

600 precursor, and when supplied to an E. coli culture expressing F3H and FLS from Arabidopsis thaliana,

601 production of garbanzol and resokaempferol was detected. Resokaempferol could be further converted into

602 fisetin by expression of F3’H genes from A. thaliana and Pn. hybrida fused with a cytochrome-P450

603 reductase (CPR) from Catharanthus roseus (Stahlhut et al. 2015). The three enzymes were then combined

604 with the isoliquiritigenin biosynthetic pathway, consisting of TAL from Rhodobacter sphaeroides, 4CL from

605 Pr. crispum, CHI from Medicago sativa, and a fusion protein of CHS from Pn. hybrida and CHR from

606 Astragalus mongholicus, to yield 0.3 mg l-1 of fisetin (Stahlhut et al. 2015). This work allowed reconstruction

607 of a complete pathway for the biosynthesis of fisetin from L-tyrosine as the starting precursor using enzymes

608 from various sources, and the identified pathway is currently being transferred to Gram-positive hosts.

609 Bioinformatics and modelling

610 There were multiple activities within BacHBerry, both within the discovery and the microorganism

611 engineering sectors, which required the use of bioinformatics tools. Hence, the mathematical modeling and

612 computational parts of the project aimed at addressing three major problems. The first two were related to

613 gene expression, either for performing functional genomics analysis using the RNA-seq data from berries or

614 to design sequences for heterologous expression of plant genes in microbes. The third problem concerned

615 metabolic network optimization for both a single population and for a community of microbes. We detail

616 below the main results obtained on each of the above activities. Other subjects that were addressed as a part

617 of this project were metabolic communication between the different species in a community (Milreu et al.

618 2014; Andrade et al. 2016) and modeling dynamic metabolic networks for potential application in strain

25

619 optimization (Costa et al. 2014; Hartmann et al. 2015a; Hartmann et al. 2015b; Costa et al. 2016; Hartmann

620 et al. 2016).

621 Sequencing data analysis and plant functional genomics

622 Understanding which genes are differentially expressed during phenolic production by plants requires the

623 capacity to undertake a rapid analysis of RNA-seq data. One of the main problems to be addressed in this

624 case is the search for coding sequences that are homologous to those already present in sequence databases,

625 which involves identification, selection and annotation of the best matches. Currently, such analyses rely

626 heavily on manual curation as well as on the manual assembly of results obtained using different

627 computational procedures and sequence databases. Automated processes become crucial in the light of the

628 ever-increasing amounts of generated transcriptome data. Furthermore, automation of processes also allows

629 improvement of the quality of the analyses, as the likelihood of human error grows with the amount of

630 manual processing and verifications necessary. Moreover, most of the existing tools are able to annotate the

631 transcriptome but lack the curation functionalities that are as important as the primary homology-based

632 identification of candidate genes.

633 As mentioned previously we developed a pipeline called MassBlast (Veríssimo et al. submitted), to improve

634 and accelerate the annotation procedure of RNA-seq data. Besides allowing for operators to query multiple

635 databases for performing homology searches, the tool also provides a set of parameters that can be defined in

636 a user-friendly way. MassBlast seeks the best alignment between the sequences in a dataset consisting of

637 contigs obtained from RNA-seq assemblies and a database of known sequences. In the context of BacHBerry

638 and as a general validation of the method, MassBlast was used for the analysis of the blackberry

639 transcriptome, and preliminary results showed a significant improvement in annotation speed (five minutes

640 versus at least 26 working days, to annotate all known CYP families in blackberry) with very high

641 accuracies. MassBlast is freely available at https://github.com/averissimo/mass-blast/.

642 Sequence design for heterologous expression

26

643 Synthetic biology aims at constructing microorganisms capable of producing economically-relevant

644 chemical products in a cost-effective way, which involves optimizing the functioning of metabolic pathways

645 and reactions. These pathways and reactions involve proteins that are frequently heterologous. In order to

646 increase the production of metabolites, it is possible to optimize heterologous protein production in the target

647 host. This may be done by altering the codons in the gene that encodes for that protein. Each codon is a

648 sequence of three nucleotides encoding one amino acid that is part of a protein. Since each amino acid can be

649 encoded by several synonymous codons, a given protein can be expressed using a large number of different

650 codon combinations.

651 To address this problem, we worked on improving the machine learning models capable of predicting protein

652 expression levels based on their codon usage frequency (Fernandes et al. 2016) by using Support Vector

653 Regression (SVR) and Partial Least Squares (PLS). SVR yields predictions that surpass those of PLS. It has

654 been shown that it is possible to improve the predictive ability of the models by using two additional input

655 features, namely codon identification number and codon count, besides the already used codon bias and

656 minimum free energy. In addition, applying ensemble averaging to the SVR or the PLS model improved the

657 results even further. Different ensembles and features can be tested with the aim of further improving the

658 prediction models.

659 Metabolic network optimization

660 Another goal of the computational activities of BacHBerry was to explore methods for in silico metabolic

661 engineering of phenolic production in L. lactis and C. glutamicum. For this we selected four model target

662 compounds: fisetin, trans-resveratrol, pelargonidin and quercetin. Additionally, optimization of malonyl-

663 CoA production was also considered, since this is known to be a major bottleneck in heterologous flavonoid

664 production. The first step involved performing network optimization for a single population, which included

665 prediction of knock-out strategies and determination of minimal sets of genetic interventions that could

666 guarantee an increased production of the target compound production based on stoichiometric models for the

667 two organisms. These predictions were compared with experimental results and model refinement was

27

668 conducted in cases of discrepancies. For the second step, we considered a use of microbial consortia for

669 production purposes. A method was developed that is being used for modeling the production of the

670 compounds of interest for BacHBerry using L. lactis and C. glutamicum. Other, possibly larger consortia,

671 may be considered in the future.

672 Single strain-based production

673 Recently, several computational methods for in silico metabolic network optimization have emerged and are

674 currently being applied in metabolic engineering (Vera et al. 2010; Zomorrodi et al. 2012; Machado et al.

675 2015; Simeonidis and Price 2015; Bergdahl et al. 2015). They are based on different approaches and

676 rationales, thereby leading to distinct solutions. One key issue is the multitude of results, known to be

677 dependent on the specific algorithms and solvers, used. In order to address this problem, a consensus-based

678 approach was developed, following our work on meta-analysis of transcriptomic studies (Caldas et al. 2014).

679 This method is based on running several optimization procedures and analyzing a posteriori the solutions,

680 looking for a consensus. The rationale is to have rankings of hypotheses that may provide confidence in

681 particular sets of proposed genetic alterations from various aspects.

682 In the case of a single population of microbes (i.e. a monoculture), the genome-scale models of L. lactis and

683 C. glutamicum were extended with the biosynthetic pathways of the four above-mentioned target compounds

684 (Figure 9). Subsequently, five different optimization methods were applied to the genome-scale models

685 (extended with the heterologous pathways): OptKnock (Burgard et al. 2003), MOMA (Segrè et al. 2002),

686 OptGene (Patil et al. 2005), RoboKod (Stanford et al. 2015), and RobustKnock (Tepper and Shlomi 2010),

687 and the consensus ranking was obtained for each strain and compound. The hypotheses were generated with

688 the rank product test and the outputs were lists of gene deletions that would achieve the best ranks using

689 defined criteria, namely maximum predicted target compound production, maximizing the minimum

690 predicted target compound production and distance from the wild-type flux distribution. These results were

691 then further filtered and experimentally validated, resulting in several promising mutant strains where

28

692 production was increased by up to three-fold. Implementation of this pipeline is possible via a software

693 package called OptPipe, available at https://github.com/AndrasHartmann/OptPipe.

694 We then explored the concept of multi-objective optimization in the field of metabolic engineering when

695 both continuous and integer decision variables were involved in the model. In particular, we proposed multi-

696 objective models to suggest reaction deletion strategies and to deal with problems where several functions

697 must be optimized simultaneously, such as maximization of bioproducts while minimizing toxicity. We thus

698 introduced Multi-Objective Mixed integer Optimization for metabolic engineering, a computational

699 framework that aims to model and solve optimization problems, generated for predicting reaction knockout

700 strategies by means of multi-objective programming. We compared the results from our method with those

701 obtained by using the well-known bi-level optimization model (OptKnock). Furthermore, we studied two

702 multi-objective optimization problems arising from the metabolic engineering of microorganisms, showing

703 that indeed a multi-objective setting is promising, as it expands the set of generated hypotheses and allows

704 inclusion of multiple goals, simultaneously.

705 For a community of microbes

706 Pure microbial cultures have long been used for production of high-value compounds, as illustrated by the

707 example of S. cerevisiae producing artemisinic acid, an important precursor for the anti-malaria drug

708 artemisinin, on a large scale (Ro et al. 2006; Lenihan et al. 2008). However, there is increasing evidence that

709 such processes could run more efficiently if a community of microbial species is used (Logan and Rabaey

710 2012; Jones et al. 2016; Wen et al. 2016). There are multiple reasons including an increased production

711 efficiency of the system due to lessening of metabolic burden for individual cells. Another important

712 consideration is that such community systems might have higher chances of avoiding toxic effects specific

713 for individual microorganisms. Microbial consortia are believed to be able to sustain more complex

714 production pathways, as well as being more robust as a group. The challenge, however, remains in

715 establishing precisely which set of strains or species is best for the production of any given compound.

29

716 As part of BacHBerry, we introduced an initial topological model and a combinatorial algorithm that enabled

717 us to propose optimal consortia to produce compounds that were either exogenous to the consortium, or were

718 endogenous but where interaction between the species in the consortium could improve the production line

719 (Julien-Laferrière et al. 2016). For initial validations of the model and the method, we applied it to two case-

720 studies taken from the literature. The first case involved production of two bioactive compounds, penicillin

721 and cephalosporin C, both of which are antibiotics used in the pharma industry. A synthetic consortium

722 composed of four different species, three Actinobacteria (Streptomyces cattleya, Rhodococcus jostii RAH_1

723 and Rc. erythropolis BG43) and one methanogenic Archaea (Methanosarcina barkeri), was then run through

724 the algorithm, allowing introduction of genetic manipulations and regulatory processes. We showed that the

725 best solution involved only two of the four species, namely St. cattleya and M. barkeri (Julien-Laferrière et

726 al. 2016).Another considered case was a consortium that may be seen as artificial in the sense that it

727 associates two wild-type populations of microbes, pathogenic γ-proteobacteria Klebsiella pneumoniae and

728 methanogenic Archaea M. mazei, which normally do not interact in nature. K. pneumoniae is a natural

729 producer of 1,3-propanediol, which is an important building block for polymer production. Our results

730 demonstrated that co-cultivation of K. pneumoniae with M. mazei may enable obtaining higher yields of this

731 compound in Klebsiella (Julien-Laferrière et al. 2016). This supported a previous study involving co-

732 cultures of Clostridium sp. with methanogenes such as Methanosarcina sp. CHTI55 (Koesnandar et al.

733 1990). The developed algorithms are currently being used for developing consortia of polyphenol-producing

734 Gram-positive bacteria. This task was inspired by the work of Jones and colleagues, who were able to

735 achieve an overall 970-fold improvement in titers for flavan-3-ols production from phenylpropanoic acids by

736 using a co-culture of two E. coli strains with further optimization of cultivation conditions, as compared to

737 previous studies that used monocoltures for this purpose (Jones et al. 2016).

738 Recovery and purification of polyphenols from fermentation broth

739 Downstream processing is an important part of any production process and consists of steps required for

740 recovery and purification of biosynthetic products from fermentation broth, which may have a significant

741 impact on the final costs of a product (Goldberg 1997). In the design of downstream processing within

30

742 BacHBerry, trans-resveratrol (Figure 7) was used as the test product, being one of the model phenolic

743 compounds that was successfully produced in both C. glutamicum and L. lactis.

744 In a typical fermentation broth, most of the components present are either hydrophilic (e.g., salts, organic

745 acids, non-consumed carbon sources (usually glucose), nitrogen source (ammonium sulfate, for instance)) or

746 are much larger molecules than trans-resveratrol (e.g., proteins, nucleic acids, cells in suspension). This

747 suggested a downstream processing design strategy that used either solubility and hydrophobicity as the

748 driving forces, or alternatively – size exclusion. Since, at low concentrations (around 100 mg l-1),

749 crystallization would involve high energy costs per kg of product recovered, this option was discarded. Two

750 possible extraction techniques, namely liquid-liquid extraction and solid-liquid adsorption, were investigated

751 as means of recovering and purifying extracellularly produced trans-resveratrol from a C. glutamicum

752 fermentation broth containing spent CGXII medium and glucose, due to the higher production efficiency

753 found in this bacterium. The downstream process design aimed at a recovery of at least 80% and a purity of

754 95% trans-resveratrol.

755 Liquid-liquid extraction is a technique to separate a given compound from a feed solution using an

756 immiscible auxiliary phase that should be selective for that compound (Cox and Rydberg 2004). The

757 logarithm (base 10) of the partition coefficient of trans-resveratrol between the aqueous and different

758 potential organic phases was determined experimentally for several different solvents (Figure 10). The spent

759 fermentation broth from C. glutamicum was used as the aqueous phase in these experiments.

760 All tested organic solvents performed very well in extracting trans-resveratrol (minimum log P value is ~0.5,

761 corresponding to a partition coefficient of 3.2), identifying them as suitable candidates for the recovery of

762 this compound (when having different impurities present) (Figure 10).

763 One of the drawbacks of using liquid-liquid extraction, however, is that it may interfere with protein folding

764 and stability (Prince and Smith 2004). Besides measuring the partition of the polyphenols of interest in the

765 aqueous and organic phases, the same analysis was performed for the total protein content. Although a

766 considerable part of the total protein remained in the aqueous phase, some of the protein seemed to be

31

767 present in between the two liquid phases (probably as a result of denaturation and foaming) (Figure 11).

768 This phenomenon might reduce the recovery of the desired phenolics, as it was also observed experimentally

769 (data not shown).

770 Solid-liquid adsorption using hydrophobic resins is another way of purifying trans-resveratrol. As in liquid-

771 liquid extraction, the high hydrophobicity of trans-resveratrol can be used to recover it selectively from the

772 fermentation broth, leaving the more hydrophilic impurities behind. After an initial resin screen, one of the

773 resins selected was the polystyrene-divinylbenzene based Amberlite XAD-16 from Sigma-Aldrich (particle

774 diameter: 250-841 µm; dipole moment: 0.3; surface area: 900 m2 g-1; pore diameter: 100 Å).

775 To test the ability of this resin to recover trans-resveratrol from C. glutamicum fermentation broth

776 (containing CGXII spent medium and glucose), a lab-scale experiment was performed with an AKTA

777 Explorer chromatographic workstation (GE Healthcare, Sweden) (see Figure 12). A 30 ml column was

778 packed with resin and pre-equilibrated with MilliQ water. Afterwards, 10 ml of sample was injected and a

779 washing step with pure water was performed for 10 column volumes (CV). During elution, a gradient of

780 water/ethanol was run for about 12 CV and the peak of trans-resveratrol showed up at around 18 CV, at an

781 ethanol percentage of 50% (v/v). Regeneration was performed with 100% ethanol.

782 More hydrophilic molecules such as organic acids, sugars and salts appeared in the flow-through (the first 10

783 CV). During the water/ethanol gradient, molecules desorbed as a function of their hydrophobicity. More

784 hydrophobic molecules, such as trans-resveratrol, are supposed to elute at higher ethanol content in the

785 mobile phase. In this case, trans-resveratrol appeared to be the compound eluting between 16 and 20 CV,

786 since it also showed absorption at 304 nm (Camont et al. 2009).

787 Although both liquid-liquid extraction and solid-liquid adsorption present their own advantages and

788 disadvantages, a strong point for using adsorption is the use of ethanol instead of any other organic solvent

789 that might not be regarded as GRAS. Moreover, not using liquid-liquid extraction avoids any possible

790 problems related with protein precipitation and emulsification. The ethanol used during elution in adsorption

32

791 needs to be recovered from water by distillation (the azeotrope poses no problem as 80% (v/v) ethanol can be

792 used).

793 The proposed downstream process started with a solid-liquid separation step by centrifugation to separate

794 biomass from the liquid broth. The clarified liquid phase was then sent continuously to one of two adsorption

795 columns, operating staggered in time. The fraction rich in trans-resveratrol was subsequently evaporated,

796 resulting in a wet slurry containing the polyphenol in the solid phase. This slurry was filtered in a rotary

797 vacuum filter and washed with water to remove remaining impurities adhered to the surface of the crystals.

798 The final step was a drying step to obtain the product in its final solid form. All the outlet streams containing

799 ethanol were directed to a distillation column in order to recover the solvent. A make-up stream ethanol was

800 added to compensate for losses.

801 Optimization of phenolic production by fermentation and scale-up of bioprocesses

802 There are multiple differences between microbial cultivation in shake flasks versus in bioreactors. The

803 former lacks monitoring and control of various parameters, such as dissolved O2 concentration and pH (Zelle

804 et al. 2010). Furthermore, the fed-batch fermentation mode, where one or more nutrient is being supplied

805 throughout cultivation (Moulton 2014), can be done in a controllable and reproducible fashion only in

806 bioreactors. These properties make bioreactors a suitable stage for optimization of cultivation conditions,

807 often leading to significant improvements in production yields (Hujanen et al. 2001; Ratnam et al. 2003; Li

808 et al. 2010; Rani and Appaiah 2011).

809 From the beginning of the project it was clear that optimization of fermenter operation would need to be

810 carried out using a model compound, because any newly-identified compounds would only be available at

811 the end of the project. Hence this section of the project was focused on optimization of production of the

812 model compound trans-resveratrol. However, it was expected that the strategies defined and the conclusions

813 extracted from these results on trans-resveratrol will be valid for the optimization of the fermentation

814 processes for the production of other target compound. Several L. lactis and C. glutamicum strains designed

815 within the project were evaluated and used for fermenter process optimization. The experiments were carried

33

816 out in agitated 2 to 5 litre fermenters with automatically controlled pH, dissolved oxygen and temperature.

817 Whenever required, oxygen was transferred to the medium by air injection at the bottom of the vessel.

818 A major goal of the project was the development of an economically feasible and sustainable industrial

819 process, therefore the following key optimization criteria were defined: (a) minimisation of the operation and

820 investment cost and (b) maximisation of the production rate and operation yield. To achieve these

821 optimization goals, the following variables have been investigated: (i) fermentation medium composition, (ii)

822 dissolved oxygen concentration, (iii) operation strategy (batch vs. fed-batch and integrated vs. separate

823 downstream operation). Our experiments suggested that there were several constraints limiting the operation

824 strategy to be implemented, namely: cell metabolism, the stability of trans-resveratrol inside the fermenter,

825 and the affinity of trans-resveratrol for the adsorption medium and solvents compared to other compounds

826 present in the fermentation broth.

827 Medium composition is an important parameter that often has a significant contribution towards the final

828 production costs. According to our initial experiments, supplementation of C. glutamicum minimal medium

829 with Brain-Heart Infusion (BHI) and a fatty-acid biosynthesis inhibitor, cerulenin, had a strong positive

830 effect on the final titers of trans-resveratrol. However, removing these two components from the medium

831 reduced medium costs by approximately 300-fold, resulting in a more than 50-fold drop in production costs

832 (267 €/g compared to 5 €/g of trans-resveratrol without supplementation of BHI and cerulenin).

833 We also evaluated the feasibility of producing phenolic compounds from waste materials, namely cheese

834 whey using L. lactis as a host. Whey is a lactose-rich dairy industry by-product with a high environmental

835 impact, if dumped untreated (Palmieri et al. 2017). To achieve this task, our production strain was modified

836 to allow it to grow and produce trans-resveratrol from lactose by introducing a plasmid carrying the lac

837 operon (Maeda and Gasson 1986). Production at 2 litre-scale was tested using lactose as carbon source and

838 comparing the productivity between the cultures with glucose or lactose. These experiments demonstrated

839 that the production of trans-resveratrol from lactose, and therefore from whey, is possible with slightly

840 inferior productivity compared to glucose. However, if whey is chosen as the fermentation medium, other,

34

841 more important, issues arise. The cost of whey is comparable to the cost of glucose but the cost of the

842 fermentation medium is only a small part of the total production costs, so variables such as whey availability

843 and quality, as well as the stability of supply and collecting logistics will determine the feasibility of the

844 operation more than the small losses in productivity. Clearly, if a phenolic production industrial unit were to

845 be implemented inside a dairy company with excess whey, the whey might then be the best raw material.

846 Another parameter investigated was the influence of dissolved oxygen concentration on the production

847 efficiency of L. lactis. This bacterium can activate its respiratory chain in the presence of hemin in the

848 culture medium, and this has been demonstrated to enhance its survival allowing it to grow to higher cell

849 densities (Duwat et al. 2001). Therefore the production strain efficiency was tested under the following

850 conditions: aerobic with hemin added to the fermentation broth, aerobic without hemin, semi-aerobic, and

851 fully anaerobic. In line with the available data, there was a significant increase in the final biomass when the

852 strain was grown under aerobic conditions with hemin. However, the opposite was observed for the

853 production of trans-resveratrol where the highest amounts of the compound were detected under anaerobic

854 conditions.

855 Lastly, we have also worked on establishing a system for continuous product removal from the fermentation

856 broth in order to simplify downstream processing and reduce trans-resveratrol oxidation due to prolonged

857 incubation. Two strategies for continuous adsorption and storage of secreted trans-resveratrol in a resin have

858 been devised and tested: (i) “in situ” product removal that consisted of adding a resin directly to the

859 fermentation broth and (ii) “continuous product removal” (ConPR) that involved constant pumping of the

860 fermentation broth through a column filled with the adsorption resin. The supernatant was separated from

861 cells via a tangential flow micro-filtration device, sent though the column, and then returned back into the

862 fermenter (Figure 13).

863 The obtained results demonstrated that both the “in situ” and ConPR strategies lowered the trans-resveratrol

864 productivity, probably due to co-adsorption of p-coumaric acid, the precursor for trans-resveratrol. This

865 effect was most pronounced in the “in situ” operation mode due to continuous contact of the resin with the

35

866 supernatant and cells. In the ConPR operation mode the effect was reduced due to low supernatant flow rate

867 through the resin, however adsorption of trans-resveratrol was also reduced. In contrast, in the “in situ”

868 mode the amount of adsorbed end-product was nearly identical to the amount of extracellular and

869 intracellular trans-resveratrol produced in control experiments without addition of resin. Although the two

870 techniques did not increase the overall productivity, they still simplified the downstream processing and may

871 lead to a production cost reduction by eliminating the need to break down the cells. However, future research

872 may provide a solution to avoid precursor co-adsorption resulting in increased polyphenol productivity.

873 Dissemination, training, societal and ethical issues

874 In addition to applying the latest technological advances from life sciences for establishing a pipeline for

875 microbial-based polyphenol production, BacHBerry analyzed the economic potential of the possible products

876 developed from this research. The assessment of economic issues of the project has been focused on the

877 market potential of the natural colorants identified during the screening of the germplasm collection. This

878 was done along with obtaining an overview of the relevant regulations and procedures for approval of novel

879 food additives, including colorants, in the EU, the United States (US), and China. Regulations within both

880 the EU and China confer a strict set of rules for bringing a novel food colorant, even a natural one, to the

881 market. These rules consider not only the composition and purity of the substance, but also the process

882 through which the compounds have been produced. In contrast, in the US, a new food additive can gain

883 access to the market by simply passing the GRAS assessment. Hence, if a novel polyphenolic colorant could

884 be produced using engineered microbial cell factories, the US market should probably be the first market to

885 be targeted, owing to less strict regulatory requirements for the approval.

886 Another topic addressed in BacHBerry was access and benefit sharing of genetic resources. As the project

887 was built upon screening of berry species from multiple countries, including China, Russia, Chile, Portugal,

888 and the UK, it is important to harness these resources in a responsible manner. The Nagoya Protocol on

889 Access to Genetic Resources and the Fair and Equitable Sharing of Benefits Arising from their Utilization

890 (NP-ABS) is an agreement consisting of a set of rules for fair sharing of monetary and non-monetary benefits

36

891 arising from the usage and commercialization of genetic resources and traditional knowledge associated with

892 those. It aims at achieving fairness and equity of gain distribution, especially towards indigenous and local

893 communities (SCBD 2011; Morgera et al. 2014). The Protocol came into force in the EU as of November

894 20th 2016. As most of the BacHBerry project partners were from EU Member States, we began collecting

895 information about the best practices suggested from both the Convention on Biological Diversity (CBD) and

896 the EU, and kept monitoring the relevant guidelines and updates from the EU on implementing ABS. Among

897 the home countries of the project partners China, Denmark, France, Germany, Switzerland, and UK ratified

898 or accepted the NP-ABS (as of November 2016) and began implementing the aforementioned regulations

899 (European Parlament 2014). The UK also made its own regulation on implementing the Protocol (European

900 Parlament 2014; NMRO (UK) 2015; Secretary of State (UK) 2015).

901 Funded under the theme of “Knowledge-based Bio-Economy”, a final aim of the project was to provide

902 scientific and technical solutions to harness berry resources, as well as to contribute to sustainable

903 development of the economy. We consequently investigated the possible contributions of our research to

904 build a more sustainable society based on environmental, economical, and social impacts (Pei and Schmidt

905 2016). Furthermore, we conducted several open dialog events, namely workshops and interviews with

906 potential stakeholders from Austria, Denmark, and China. One of the surprising outcomes was that the

907 general public raised concerns over the use of genetically-modified organisms (GMOs) in the pipeline a lot

908 less frequently than originally expected. A free, science-based game for mobile devices called “Berry

909 Maker” was produced (http://www.berrymaker.com). The game allows the user to follow the pipeline

910 designed within our project, from acquisition of diverse berries, to extraction and analysis of biosynthetic

911 genes, to microbial fermentation-based production of specific polyphenols and running a biotech factory that

912 is exposed to real world situations, such as price fluctuations. The game is also available on iTunes1 and

913 Google Play2 store. Lastly, a 22 minute documentary film was produced, showing sampling of berries,

1 See: https://itunes.apple.com/us/app/berrymaker-dna-soda-factory/id1163115116?ls=1&mt=8

2 See: https://play.google.com/store/apps/details?id=com.biofaction.berrymaker

37

914 analysis of interesting compounds, and experiments with natural color pigments for food products from

915 across Europe (see: https://vimeo.com/193467652).

916 Conclusions

917 During three years of the project, the BacHBerry partners successfully established a complete pipeline for

918 screening, bioprospecting, identification, and microbial production of plant phenolic compounds in Gram-

919 positive bacteria. A summary of the most important results is presented in Table 5. The major highlights are

920 construction of the extensive germplasm collection database, establishment of a method for high-throughput

921 screening of bioactive extracts using the yeast-based human disease models, generation of a vast berry

922 transcriptome database, construction of microbial cell factories in L. lactis and C. glutamicum for the

923 production of polyphenols with yields comparable to those currently available from the literature, release of

924 several software packages for transcriptome analysis and in silico-based metabolic engineering, as well as

925 optimization of fermentation conditions and downstream processing for the selected organisms aimed at

926 increasing the yield and the purify of the final products. The pipeline has been completely validated and is

927 available to be picked up by the biotech industry for further improvement and commercialization. The

928 developed methods and protocols should be suitable for discovery, as well as production of most groups of

929 polyphenols in the above-mentioned organisms, without a need for significant alterations and adaptations.

930 Overall, this project provided the scientific community with enabling tools and resources to further

931 advancing research on berries, phenolic compounds, and metabolic engineering of microorganisms.

932 Additional efforts were also focused on communication with stakeholders, public outreach, and

933 popularization of science among the younger generation. The majority of the project outcomes is already

934 available to the scientific community and the industry in form of peer-reviewed articles, or will be published

935 within the coming year.

936

937 Acknowledgments

38

938 The authors would like to thank the European Union’s Seventh Framework Programme (BacHBerry, Project

939 No. FP7-613793, and FP7-PEOPLE-2013-COFUND, Project No. FP7-609405) for the financial support.

940 AD, RF, MHHN, PG, SGS, and JF would also like to acknowledge the Novo Nordisk Foundation. We

941 express our gratitude to Dr. Martha Cyert (Stanford School of Medicine, EUA), Dr. Hitoshi Shimoi (National

942 Research Institute of Brewing, Japan) and Dr. Yoshio Araki (Graduate School of Biosphere Science,

943 Hiroshima University, Japan) for providing the yeast strain YAA5. We also thank Dr. Ian Macraedie, RMIT

944 University, Australia) for providing the plasmid p416_GPDpr-GFP-A42.

945

946 Abbreviations

947 4CL, 4-coumaryl-CoA ligase; AD, Alzheimer's Disease; ANS, anthocyanidin synthase; ALS, amyotrophic

948 lateral sclerosis; anthocyanidin synthase; ANR, anthocyanidin reductase; C3’H, p-coumaroyl

949 shikimate/quinate 3'-hydroxylase; C4H, cinnamate 4-hydroxylase; CBD, Convention on Biological

950 Diversity; CaN, calcineurin; CDRE, Calcineurin-Dependent Responsive Element; CHI, chalcone isomerase;

951 CHR, chalcone reductase; CHS, chalcone synthase; CPR, cytochrome-P450 reductase; CVs, column

952 volumes; CYP, cytochromes P450; DFR, dihydroflavonol 4-reductase; F3H, flavanone 3-hydroxylase; F3’H,

953 flavonoid 3’-hydroxylase; F3’5’H, flavonoid 3’-5’-hydroxylase; FLS, flavonol synthase; FNS, flavone

954 synthase; FP7, European Commission’s the Seventh Framework programme; GFP, Green Fluorescent

955 Protein; GRAS, generally regarded as safe; HCT, hydroxycinnamoyl-CoA shikimate/quinate

956 hydroxycinnamoyltransferase; HD, Huntington's Disease; HPLC, high-performance liquid chromatography;

957 LAR, leucoanthocyanidin reductase; LC-MS, liquid-chromatography mass-spectrometry; NDs,

958 Neurodegenerative diseases; NFAT, Nuclear Factor of Activated T-cells; NFκB, Nuclear Factor κB; NP-

959 ABS, The Nagoya protocol on access to genetic resources and the fair and equitable sharing of benefits

960 arising from their utilization; OMT, O-methyltransferase; PAL, phenylalanine ammonia-lyase; PCA,

961 principal component analysis; PD, Parkinson's Disease; PLS, partial least squares; RNA-seq, RNA

39

962 sequencing technology; STS, stilbene synthase; SVR, support vector regression; TAL, tyrosine ammonia-

963 lyase; UFGT, flavonoid 3-O-glucosyltransferase.

964

965 Appendices

966 Supplementary_Materials.docx

967 Supplementary_Tables.docx

968

969 References

970 Andrade R, Wannagat M, Klein CC, et al (2016) Enumeration of minimal stoichiometric precursor sets in

971 metabolic networks. Algorithms Mol Biol 11:25. doi: 10.1186/s13015-016-0087-3

972 Aranaz I, Acosta N, Mengíbar MN, et al (2016) InFiQuS: Making the Best of Leftovers. In: Domı́nguez de

973 Marı́a P (ed) Industrial biorenewables : a practical viewpoint. John Wiley & Sons, Inc., Hoboken, US,

974 pp 363–364

975 Bak S, Olsen CE, Halkier BA, Møller BL (2000) Transgenic tobacco and Arabidopsis plants expressing the

976 two multifunctional sorghum cytochrome P450 enzymes, CYP79A1 and CYP71E1, are cyanogenic and

977 accumulate metabolites derived from intermediates in Dhurrin biosynthesis. Plant Physiol 123:1437–

978 48. doi: 10.1104/pp.123.4.1437

979 Becker J, Wittmann C (2012) Bio-based production of chemicals, materials and fuels – Corynebacterium

980 glutamicum as versatile cell factory. Curr Opin Biotechnol 23:631–640. doi:

981 10.1016/j.copbio.2011.11.012

982 Becker J, Armstrong G, Vandermerwe M, et al (2003) Metabolic engineering of for the synthesis of the

40

983 wine-related antioxidant resveratrol. FEMS Yeast Res 4:79–85. doi: 10.1016/S1567-1356(03)00157-0

984 Beekwilder J, Wolswinkel R, Jonker H, et al (2006) Production of resveratrol in recombinant

985 microorganisms. Appl Environ Microbiol 72:5670–5672. doi: 10.1128/AEM.00609-06

986 Bergdahl B, Sonnenschein N, Machado D, et al (2015) Genome-Scale Models. In: Villadsen J (ed)

987 Fundamental Bioengineering. Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim, Germany, pp 143–

988 182

989 Bhan N, Xu P, Khalidi O, Koffas MAG (2013) Redirecting carbon flux into malonyl-CoA to improve

990 resveratrol titers: Proof of concept for genetic interventions predicted by OptForce computational

991 framework. Chem Eng Sci 103:109–114. doi: 10.1016/j.ces.2012.10.009

992 Bharadwaj P, Martins R, Macreadie I (2010) Yeast as a model for studying Alzheimer’s disease. FEMS

993 Yeast Res 10:961–969. doi: 10.1111/j.1567-1364.2010.00658.x

994 Blomstedt CK, Gleadow RM, O’Donnell N, et al (2012) A combined biochemical screen and TILLING

995 approach identifies mutations in Sorghum bicolor L. Moench resulting in acyanogenic forage

996 production. Plant Biotechnol J 10:54–66. doi: 10.1111/j.1467-7652.2011.00646.x

997 Bodvard K, Jörhov A, Blomberg A, et al (2013) The Yeast Transcription Factor Crz1 Is Activated by Light

998 in a Ca2+/Calcineurin-Dependent and PKA-Independent Manner. PLoS One 8:e53404. doi:

999 10.1371/journal.pone.0053404

1000 Borrill P, Ramirez-Gonzalez R, Uauy C (2016) expVIP: a Customizable RNA-seq Data Analysis and

1001 Visualization Platform. Plant Physiol 170:2172–86. doi: 10.1104/pp.15.01667

1002 Burgard AP, Pharkya P, Maranas CD (2003) Optknock: A bilevel programming framework for identifying

1003 gene knockout strategies for microbial strain optimization. Biotechnol Bioeng 84:647–657. doi:

1004 10.1002/bit.10803

41

1005 Burkovski A (ed) (2008) Corynebacteria: Genomics and Molecular Biology. Caister Academic Press

1006 Caldas J, Vinga S, Nordmann A, et al (2014) Global Meta-Analysis of Transcriptomics Studies. PLoS One

1007 9:e89318. doi: 10.1371/journal.pone.0089318

1008 Camont L, Cottart C-H, Rhayem Y, et al (2009) Simple spectrophotometric assessment of the trans-/cis-

1009 resveratrol ratio in aqueous solutions. Anal Chim Acta 634:121–128. doi: 10.1016/j.aca.2008.12.003

1010 Cao W, Ma W, Zhang B, et al (2016) Improved pinocembrin production in Escherichia coli by engineering

1011 fatty acid synthesis. J Ind Microbiol Biotechnol 43:557–566. doi: 10.1007/s10295-015-1725-3

1012 Carvalho E, Franceschi P, Feller A, et al (2013) A targeted metabolomics approach to understand differences

1013 in flavonoid biosynthesis in red and yellow raspberries. Plant Physiol Biochem PPB 72:79–86. doi:

1014 10.1016/j.plaphy.2013.04.001

1015 Catalgol B, Batirel S, Taga Y, Ozer NK (2012) Resveratrol: French Paradox Revisited. Front Pharmacol

1016 3:141. doi: 10.3389/fphar.2012.00141

1017 Chaovanalikit A, Thompson MM, Wrolstad RE (2004) Characterization and Quantification of Anthocyanins

1018 and Polyphenolics in Blue Honeysuckle (Lonicera caerulea L.). J Agric Food Chem 52:848–852. doi:

1019 10.1021/JF030509O

1020 Chen X, Zhou L, Tian K, et al (2013) Metabolic engineering of Escherichia coli: A sustainable industrial

1021 platform for bio-based chemical production. Biotechnol Adv 31:1200–1223. doi:

1022 10.1016/j.biotechadv.2013.02.009

1023 Choi O, Wu C-Z, Kang SY, et al (2011) Biosynthesis of plant-specific phenylpropanoids by construction of

1024 an artificial biosynthetic pathway in Escherichia coli. J Ind Microbiol Biotechnol 38:1657–1665. doi:

1025 10.1007/s10295-011-0954-3

1026 Costa RS, Hartmann A, Vinga S (2016) Kinetic modeling of cell metabolism for microbial production. J

42

1027 Biotechnol 219:126–141. doi: 10.1016/j.jbiotec.2015.12.023

1028 Costa RS, Veríssimo A, Vinga S, et al (2014) Ki MoSys: a web-based repository of experimental data for

1029 KInetic MOdels of biological SYStems. BMC Syst Biol 8:85. doi: 10.1186/s12918-014-0085-3

1030 Cox M, Rydberg J (2004) Introduction to Solvent Extraction. In: Rydberg J, Cox M, Musikas C, Choppin

1031 GR (eds) Solvent extraction principles and practice, 2nd edn. Marcel Decker, Inc., New York, USA,

1032 Cragg GM, Newman DJ (2013) Natural products: A continuing source of novel drug leads. Biochim Biophys

1033 Acta - Gen Subj 1830:3670–3695. doi: 10.1016/j.bbagen.2013.02.008

1034 Dobson R, Gray V, Rumbold K (2012) Microbial utilization of crude glycerol for the production of value-

1035 added products. J Ind Microbiol Biotechnol 39:217–226. doi: 10.1007/s10295-011-1038-0

1036 Duarte K, Rocha-Santos TAP, Freitas AC, Duarte AC (2012) Analytical techniques for discovery of

1037 bioactive compounds from marine fungi. TrAC Trends Anal Chem 34:97–110. doi:

1038 10.1016/j.trac.2011.10.014

1039 Dumont FJ (2000) FK506, an immunosuppressant targeting calcineurin function. Curr Med Chem 7:731–48.

1040 Duwat P, Sourice S, Cesselin B, et al (2001) Respiration capacity of the fermenting bacterium Lactococcus

1041 lactis and its positive effects on growth and survival. J Bacteriol 183:4509–16. doi:

1042 10.1128/JB.183.15.4509-4516.2001

1043 Dvora H, Koffas MAG (2013) Microbial production of flavonoids and terpenoids. In: McNeil B, Archer D,

1044 Giavasis I, Harvey L (eds) Microbial Production of Food Ingredients, Enzymes and Nutraceuticals.

1045 Woodhead Publishing, Oxford, UK, pp 234–261

1046 Eggeling L, Bott M (eds) (2005) Handbook of Corynebacterium glutamicum. CRC Press, Boca Raton, US

1047 European Parlament (2014) REGULATION (EU) No 511/2014 OF THE EUROPEAN PARLIAMENT

1048 AND OF THE COUNCIL of 16 April 2014 on compliance measures for users from the Nagoya

43

1049 Protocol on Access to Genetic Resources and the Fair and Equitable Sharing of Benefits Arising from

1050 their Utilizati.

1051 Falcone Ferreyra ML, Rius SP, Casati P (2012) Flavonoids: biosynthesis, biological functions, and

1052 biotechnological applications. Front Plant Sci 3:222. doi: 10.3389/fpls.2012.00222

1053 Fernandes A, Vinga S, Kern A, et al (2016) Improving Protein Expression Prediction Using Extra Features

1054 and Ensemble Averaging. PLoS One 11:e0150369. doi: 10.1371/journal.pone.0150369

1055 Figueira I, Menezes R, Macedo D, et al Polyphenols beyond barriers: a glimpse into the brain.

1056 Fredes C, Yousef GG, Robert P, et al (2014) Anthocyanin profiling of wild maqui berries ( Aristotelia

1057 chilensis [Mol.] Stuntz) from different geographical regions in Chile. J Sci Food Agric 94:2639–2648.

1058 doi: 10.1002/jsfa.6602

1059 Frelet-Barrand A, Boutigny S, Moyet L, et al (2010) Lactococcus lactis, an Alternative System for

1060 Functional Expression of Peripheral and Intrinsic Arabidopsis Membrane Proteins. PLoS One 5:e8746.

1061 doi: 10.1371/journal.pone.0008746

1062 Funakoshi-Tago M, Nakamura K, Tago K, et al (2011) Anti-inflammatory activity of structurally related

1063 flavonoids, Apigenin, Luteolin and Fisetin. Int Immunopharmacol 11:1150–1159. doi:

1064 10.1016/j.intimp.2011.03.012

1065 Garcia G, Santos CN do, Menezes R (2016) High-Throughput Yeast-Based Reporter Assay to Identify

1066 Compounds with Anti-inflammatory Potential. In: Matthiesen R (ed) Proteostasis. Springer, New York,

1067 USA, pp 441–452

1068 Gaspar P, Carvalho AL, Vinga S, et al (2013) From physiology to systems metabolic engineering for the

1069 production of biochemicals by lactic acid bacteria. Biotechnol Adv 31:764–88. doi:

1070 10.1016/j.biotechadv.2013.03.011

44

1071 Ghosh D, Scheepens A (2009) Vascular action of polyphenols. Mol Nutr Food Res 53:322–331. doi:

1072 10.1002/mnfr.200800182

1073 Giusti MM, Wrolstad RE (2003) Acylated anthocyanins from edible sources and their applications in food

1074 systems. Biochem Eng J 14:217–225. doi: 10.1016/S1369-703X(02)00221-8

1075 Goldberg E (1997) Handbook of Downstream Processing. Springer Netherlands, London, UK

1076 Goszcz K, Deakin SJ, Duthie GG, et al (2015) Antioxidants in Cardiovascular Therapy: Panacea or False

1077 Hope? Front Cardiovasc Med 2:29. doi: 10.3389/fcvm.2015.00029

1078 Grand View Research Inc. (2016) Polyphenols Market Analysis By Product (Grape Seed, Green Tea,

1079 Apple), By Application (Functional Food, Functional Beverages, Dietary Supplements) And Segment

1080 Forecasts To 2024.

1081 Halvorsen BL, Carlsen MH, Phillips KM, et al (2006) Content of redox-active compounds (ie, antioxidants)

1082 in foods consumed in the United States. Am J Clin Nutr 84:95–135.

1083 Hartmann U, Sagasser M, Mehrtens F, et al (2005) Differential combinatorial interactions of cis-acting

1084 elements recognized by R2R3-MYB, BZIP, and BHLH factors control light-responsive and tissue-

1085 specific activation of phenylpropanoid biosynthesis genes. Plant Mol Biol 57:155–171. doi:

1086 10.1007/s11103-004-6910-0

1087 Hartmann A, Lemos JM, Costa RS, et al (2015a) Identification of switched ARX models via convex

1088 optimization and expectation maximization. J Process Control 28:9–16. doi:

1089 10.1016/j.jprocont.2015.02.003

1090 Hartmann A, Lemos JM, Vinga S (2015b) Modeling multiple experiments using regularized optimization: A

1091 case study on bacterial glucose utilization dynamics. Comput Biol Med 63:301–309. doi:

1092 10.1016/j.compbiomed.2014.08.027

45

1093 Hartmann A, Neves AR, Lemos JM, Vinga S (2016) Identification and automatic segmentation of

1094 multiphasic cell growth using a linear hybrid model. Math Biosci 279:83–89. doi:

1095 10.1016/j.mbs.2016.06.013

1096 Heldt H-W, Piechulla B (2011) Plant Biochemistry, 4th edn. Elsevier, London, UK

1097 Hellströ m JK, Tö rrö nen AR, Mattila PH (2009) Proanthocyanidins in Common Food Products of Plant

1098 Origin. J Agric Food Chem 57:7899–7906. doi: 10.1021/jf901434d

1099 Hernández I, Molenaar D, Beekwilder J, et al (2007) Expression of plant flavor genes in Lactococcus lactis.

1100 Appl Environ Microbiol 73:1544–52. doi: 10.1128/AEM.01870-06

1101 Herrmann K, Nagel CW (1989) Occurrence and content of hydroxycinnamic and hydroxybenzoic acid

1102 compounds in foods. Crit Rev Food Sci Nutr 28:315–347. doi: 10.1080/10408398909527504

1103 Hujanen M, Linko S, Linko Y-Y, Leisola M (2001) Optimisation of media and cultivation conditions for L

1104 (+)( S )-lactic acid production by Lactobacillus casei NRRL B-441. Appl Microbiol Biotechnol

1105 56:126–130. doi: 10.1007/s002530000501

1106 Häkkinen SH, Kärenlampi SO, Heinonen IM, et al (1999) Content of the Flavonols Quercetin, Myricetin,

1107 and Kaempferol in 25 Edible Berries. J Agric Food Chem 47:2274–2279. doi: 10.1021/JF9811065

1108 Jain N, Ramawat KG (2013) Nutraceuticals and Antioxidants in Prevention of Diseases. In: Ramawat KG,

1109 Merillon J-M (eds) Natural Products. Springer Berlin Heidelberg, Berlin, Germany, pp 2559–2580

1110 Jash SK, Mondal S (2014) Bioactive flavonoid fisetin – a molecule of pharmacological interest. Signpost

1111 Open Access J Org Biomol Chem 2:89–128.

1112 Jeandet P, Bessis R, Gautheron B (1991) The Production of Resveratrol (3,5,4’-trihydroxystilbene) by Grape

1113 Berries in Different Developmental Stages. Am J Enol Vitic 42:41–46.

1114 Jendresen CB, Stahlhut SG, Li M, et al (2015) Highly Active and Specific Tyrosine Ammonia-Lyases from

46

1115 Diverse Origins Enable Enhanced Production of Aromatic Compounds in Bacteria and Saccharomyces

1116 cerevisiae. Appl Environ Microbiol 81:4458–76. doi: 10.1128/AEM.00405-15

1117 Jojima T, Inui M, Yukawa H (2013) Biorefinery Applications of Corynebacterium glutamicum. In: Yukawa

1118 H, Inui M (eds) Corynebacterium glutamicum: Biology and Biotechnology. Springer, Berlin, Germany,

1119 pp 149–172

1120 Jones JA, Vernacchio VR, Sinkoe AL, et al (2016) Experimental and computational optimization of an

1121 Escherichia coli co-culture for the efficient production of flavonoids. Metab Eng 35:55–63. doi:

1122 10.1016/j.ymben.2016.01.006

1123 Josuttis M, Verrall S, Stewart D, et al (2013) Genetic and environmental effects on tannin composition in

1124 strawberry (Fragaria × ananassa) cultivars grown in different European locations. J Agric Food Chem

1125 61:790–800. doi: 10.1021/jf303725g

1126 Ju S, Tardiff DF, Han H, et al (2011) A Yeast Model of FUS/TLS-Dependent Cytotoxicity. PLoS Biol

1127 9:e1001052. doi: 10.1371/journal.pbio.1001052

1128 Julien-Laferrière A, Bulteau L, Parrot D, et al (2016) A Combinatorial Algorithm for Microbial Consortia

1129 Synthetic Design. Sci Rep 6:29182. doi: 10.1038/srep29182

1130 Kallscheuer N, Vogt M, Bott M, Marienhagen J (2017) Functional expression of plant-derived O-

1131 methyltransferase, flavanone 3-hydroxylase, and flavonol synthase in Corynebacterium glutamicum for

1132 production of pterostilbene, kaempferol, and quercetin. J Biotechnol. doi:

1133 10.1016/j.jbiotec.2017.01.006

1134 Kallscheuer N, Vogt M, Kappelmann J, et al (2016a) Identification of the phd gene cluster responsible for

1135 phenylpropanoid utilization in Corynebacterium glutamicum. Appl Microbiol Biotechnol 100:1871–

1136 1881. doi: 10.1007/s00253-015-7165-1

1137 Kallscheuer N, Vogt M, Marienhagen J (2016b) A Novel Synthetic Pathway Enables Microbial Production

47

1138 of Polyphenols Independent from the Endogenous Aromatic Amino Acid Metabolism. ACS Synth Biol

1139 acssynbio.6b00291. doi: 10.1021/acssynbio.6b00291

1140 Kallscheuer N, Vogt M, Stenzel A, et al (2016c) Construction of a Corynebacterium glutamicum platform

1141 strain for the production of stilbenes and (2S)-flavanones. Metab Eng 38:47–55. doi:

1142 10.1016/j.ymben.2016.06.003

1143 Kasiotis KM, Pratsinis H, Kletsas D, Haroutounian SA (2013) Resveratrol and related stilbenes: their anti-

1144 aging and anti-angiogenic properties. Food Chem Toxicol 61:112–120. doi: 10.1016/j.fct.2013.03.038

1145 Katz M, Durhuus T, Smits HP, FÖRSTER J (2011) Production of metabolites.

1146 Kim HJ, Chang EJ, Cho SH, et al (2002) Antioxidative Activity of Resveratrol and Its Derivatives Isolated

1147 from Seeds of Paeonia lactiflora. Biosci Biotechnol Biochem 66:1990–1993. doi: 10.1271/bbb.66.1990

1148 Koesnandar A, Nishio N, Nagai S (1990) Stimulation by cysteine on growth of Clostridium thermoaceticum

1149 in minimal medium. Appl Microbiol Biotechnol 32:711–714. doi: 10.1007/BF00164746

1150 Kortmann M, Kuhl V, Klaffl S, Bott M (2015) A chromosomally encoded T7 RNA polymerase-dependent

1151 gene expression system for Corynebacterium glutamicum: construction and comparative evaluation at

1152 the single-cell level. Microb Biotechnol 8:253–265. doi: 10.1111/1751-7915.12236

1153 Kritzer JA, Hamamichi S, McCaffery JM, et al (2009) Rapid selection of cyclic peptides that reduce α-

1154 synuclein toxicity in yeast and animal models. Nat Chem Biol 5:655–663. doi: 10.1038/nchembio.193

1155 Krobitsch S, Lindquist S (2000) Aggregation of huntingtin in yeast varies with the length of the

1156 polyglutamine expansion and the expression of chaperone proteins. Proc Natl Acad Sci U S A

1157 97:1589–94. doi: 10.1073/PNAS.97.4.1589

1158 Krömer JO, Sorgenfrei O, Klopprogge K, et al (2004) In-depth profiling of lysine-producing

1159 Corynebacterium glutamicum by combined analysis of the transcriptome, metabolome, and fluxome. J

48

1160 Bacteriol 186:1769–84. doi: 10.1128/JB.186.6.1769-1784.2004

1161 Kuipers OP, Beerthuyzen MM, Siezen RJ, et al (1993) Characterization of the nisin gene cluster

1162 nisABTCIPR of Lactococcus lactis. Requirement of expression of the nisA and nisI genes for

1163 development of immunity. Eur J Biochem 216:281–291. doi: 10.1111/j.1432-1033.1993.tb18143.x

1164 Kuipers OP, de Ruyter PGG., Kleerebezem M, de Vos WM (1998) Quorum sensing-controlled gene

1165 expression in lactic acid bacteria. J Biotechnol 64:15–21. doi: 10.1016/S0168-1656(98)00100-X

1166 Kula M, Majdan M, Głód D, Krauze-Baranowska M (2016) Phenolic composition of fruits from different

1167 cultivars of red and black raspberries grown in Poland. J Food Compos Anal 52:74–82. doi:

1168 10.1016/j.jfca.2016.08.003

1169 Kunji ERS, Chan KW, Slotboom DJ, et al (2005) Eukaryotic membrane protein overproduction in

1170 Lactococcus lactis. Curr Opin Biotechnol 16:546–551. doi: 10.1016/j.copbio.2005.08.006

1171 Kwiatkowski TJ, Bosco DA, LeClerc AL, et al (2009) Mutations in the FUS/TLS Gene on Chromosome 16

1172 Cause Familial Amyotrophic Lateral Sclerosis. Science 323:1205–1208. doi: 10.1126/science.1166066

1173 Kårlund A, Moor U, Sandell M, Karjalainen R (2014) The Impact of Harvesting, Storage and Processing

1174 Factors on Health-Promoting Phytochemicals in Berries and Fruits. Processes 2:596–624. doi:

1175 10.3390/pr2030596

1176 Lahtinen S, Ouwehand AC, Salminen S, von Wright A (eds) (2011) Lactic Acid Bacteria: Microbiological

1177 and Functional Aspects, 4th edn. CRC Press, Boca Raton, USA

1178 Landete JM (2011) Ellagitannins, ellagic acid and their derived metabolites: A review about source,

1179 metabolism, functions and health. Food Res Int 44:1150–1160. doi: 10.1016/j.foodres.2011.04.027

1180 Lenihan JR, Tsuruta H, Diola D, et al (2008) Developing an industrial artemisinic acid fermentation process

1181 to support the cost-effective production of antimalarial artemisinin-based combination therapies.

49

1182 Biotechnol Prog 24:1026–1032. doi: 10.1002/btpr.27

1183 Li H, Qiu T, Huang G, et al (2010) Production of gamma-aminobutyric acid by Lactobacillus brevis

1184 NCL912 using fed-batch fermentation. Microb Cell Fact 9:85. doi: 10.1186/1475-2859-9-85

1185 Li X, Sun H, Pei J, et al (2012) De novo sequencing and comparative analysis of the blueberry transcriptome

1186 to discover putative genes related to antioxidants. Gene 511:54–61. doi: 10.1016/j.gene.2012.09.021

1187 Linares DM, Kok J, Poolman B (2010) Genome sequences of Lactococcus lactis MG1363 (revised) and

1188 NZ9000 and comparative physiological studies. J Bacteriol 192:5806–12. doi: 10.1128/JB.00533-10

1189 Liu Y-B, Long M-X, Yin Y-J, et al (2013) Physiological roles of mycothiol in detoxification and tolerance to

1190 multiple poisonous chemicals in Corynebacterium glutamicum. Arch Microbiol 195:419–429. doi:

1191 10.1007/s00203-013-0889-3

1192 Logan BE, Rabaey K (2012) Conversion of Wastes into Bioelectricity and Chemicals by Using Microbial

1193 Electrochemical Technologies. Science 337:686–690. doi: 10.1126/science.1217412

1194 Luo J, Nishiyama Y, Fuell C, et al (2007) Convergent evolution in the BAHD family of acyl transferases:

1195 identification and characterization of anthocyanin acyl transferases from Arabidopsis thaliana. Plant J

1196 50:678–695. doi: 10.1111/j.1365-313X.2007.03079.x

1197 MacDonald MJ, D’Cunha GB (2007) A modern view of phenylalanine ammonia lyase. Biochem Cell Biol

1198 85:273–282. doi: 10.1139/O07-018

1199 Machado D, Herrgård MJ, Rocha I (2015) Modeling the Contribution of Allosteric Regulation for Flux

1200 Control in the Central Carbon Metabolism of E. coli. Front Bioeng Biotechnol 3:154. doi:

1201 10.3389/fbioe.2015.00154

1202 Maeda S, Gasson MJ (1986) Cloning, expression and location of the Streptococcus lactis gene for phospho-

1203 β-d-galactosidase. Microbiology 132:331–340. doi: 10.1099/00221287-132-2-331

50

1204 Maher P (2009) Modulation of multiple pathways involved in the maintenance of neuronal function during

1205 aging by fisetin. Genes Nutr 4:297–307. doi: 10.1007/s12263-009-0142-5

1206 Maher P, Dargusch R, Ehren JL, et al (2011) Fisetin Lowers Methylglyoxal Dependent Protein Glycation

1207 and Limits the Complications of Diabetes. PLoS One 6:e21226. doi: 10.1371/journal.pone.0021226

1208 Manganaris GA, Goulas V, Vicente AR, Terry LA (2014) Berry antioxidants: small fruits providing large

1209 benefits. J Sci Food Agric 94:825–833. doi: 10.1002/jsfa.6432

1210 Marhuenda J, Alemán MD, Gironés-Vilaplana A, et al (2016) Phenolic Composition, Antioxidant Activity,

1211 and In Vitro Availability of Four Different Berries. J Chem 2016:1–7. doi: 10.1155/2016/5194901

1212 Marienhagen J, Bott M (2013) Metabolic engineering of microorganisms for the synthesis of plant natural

1213 products. J Biotechnol 163:166–178. doi: 10.1016/j.jbiotec.2012.06.001

1214 Mazur SP, Sønsteby A, Nes A, et al (2014) Effects of Post-Flowering Environmental Variation along an

1215 Altitudinal Gradient on Chemical Composition of “Glen Ample” Red Raspberry (Rubus idaeus L.). Eur

1216 J Hortic Sci 79:267–277.

1217 McDougall G, Martinussen I, Stewart D (2008) Towards fruitful metabolomics: High throughput analyses of

1218 polyphenol composition in berries using direct infusion mass spectrometry. J Chromatogr B 871:362–

1219 369. doi: 10.1016/j.jchromb.2008.06.032

1220 Menezes R, Tenreiro S, Macedo D, et al (2015) From the baker to the bedside: yeast models of Parkinson’s

1221 disease. Microb Cell 2:262–279. doi: 10.15698/mic2015.08.219

1222 Miladinović B, Kostić M, Šavikin K, et al (2014) Chemical Profile and Antioxidative and Antimicrobial

1223 Activity of Juices and Extracts of 4 Black Currants Varieties ( Ribes nigrum L.). J Food Sci 79:C301–

1224 C309. doi: 10.1111/1750-3841.12364

1225 Milivojević J, Maksimović V, Nikolić M, et al (2011) Chemical and antioxidant properties of cultivated and

51

1226 wild Fragaria and Rubus berries. J Food Qual 34:1–9. doi: 10.1111/j.1745-4557.2010.00360.x

1227 Miller-Fleming L, Giorgini F, Outeiro TF (2008) Yeast as a model for studying human neurodegenerative

1228 disorders. Biotechnol J 3:325–338. doi: 10.1002/biot.200700217

1229 Milreu PV, Klein CC, Cottret L, et al (2014) Telling metabolic stories to explore metabolomics data: a case

1230 study on the yeast response to cadmium exposure. Bioinformatics 30:61–70. doi:

1231 10.1093/bioinformatics/btt597

1232 Morgera E, Tsioumani E, Buck M (2014) Unraveling the Nagoya Protocol. Brill, Leiden, The Netherlands

1233 Moulton GG (2014) Fed-Batch Fermentation. Woodhead Publishing, Cambridge, UK

1234 Moyer RA, Hummer KE, Finn CE, et al (2001) Anthocyanins, Phenolics, and Antioxidant Capacity in

1235 Diverse Small Fruits: Vaccinium, Rubus, and Ribes. J Agric Food Chem 50:519–525. doi:

1236 10.1021/JF011062R

1237 Määttä KR, Kamal-Eldin A, Törrönen AR (2003) High-Performance Liquid Chromatography (HPLC)

1238 Analysis of Phenolic Compounds in Berries with Diode Array and Electrospray Ionization Mass

1239 Spectrometric (MS) Detection: Ribes Species. J Agric Food Chem 51:6736–6744. doi:

1240 10.1021/JF0347517

1241 Määttä KR, Kamal-Eldin A, Törrönen R (2001) Phenolic Compounds in Berries of Black, Red, Green, and

1242 White Currants ( Ribes sp.). Antioxid Redox Signal 3:981–993. doi: 10.1089/152308601317203521

1243 Neves AR, Pool WA, Kok J, et al (2005) Overview on sugar metabolism and its control in Lactococcus lactis

1244 — The input from in vivo NMR. FEMS Microbiol Rev 29:531–554. doi:

1245 http://dx.doi.org/10.1016/j.fmrre.2005.04.005

1246 NMRO (UK) (2015) The Nagoya protocol on access and benefit sharing of genetic resources: compliance

1247 and guidance.

52

1248 O’Brien RJ, Wong PC (2011) Amyloid precursor protein processing and Alzheimer’s disease. Annu Rev

1249 Neurosci 34:185–204. doi: 10.1146/annurev-neuro-061010-113613

1250 Oliveira AP, Nielsen J, Förster J (2005) Modeling Lactococcus lactis using a genome-scale flux model.

1251 BMC Microbiol 5:39. doi: 10.1186/1471-2180-5-39

1252 Outeiro TF, Lindquist S (2003) Yeast cells provide insight into alpha-synuclein biology and pathobiology.

1253 Science 302:1772–5. doi: 10.1126/science.1090439

1254 Palmieri N, Forleo MB, Salimei E (2017) Environmental impacts of a dairy cheese chain including whey

1255 feeding: An Italian case study. J Clean Prod 140:881–889. doi: 10.1016/j.jclepro.2016.06.185

1256 Pan M-G, Xiong Y, Chen F (2013) NFAT gene family in inflammation and cancer. Curr Mol Med 13:543–

1257 54.

1258 Pandey RP, Parajuli P, Koffas MAG, Sohng JK (2016) Microbial production of natural and non-natural

1259 flavonoids: Pathway engineering, directed evolution and systems/synthetic biology. Biotechnol Adv

1260 34:634–662. doi: 10.1016/j.biotechadv.2016.02.012

1261 Paredes-López O, Cervantes-Ceja ML, Vigna-Pérez M, Hernández-Pérez TT (2010) Berries: improving

1262 human health and healthy aging, and promoting quality life - a review. Plant Foods Hum Nutr 65:299–

1263 308. doi: 10.1007/s11130-010-0177-1

1264 Pátek M, Nešvera J (2013) Promoters and Plasmid Vectors of Corynebacterium glutamicum. In: Yukawa H,

1265 Inui M (eds) Corynebacterium glutamicum: Biology and Biotechnology, 23rd edn. Springer, Berlin,

1266 Germany, pp 51–88

1267 Patil K, Rocha I, Förster J, et al (2005) Evolutionary programming as a platform for in silico metabolic

1268 engineering. BMC Bioinformatics 6:308. doi: 10.1186/1471-2105-6-308

1269 Pauli GF, Chen S-N, Friesen JB, et al (2012) Analysis and Purification of Bioactive Natural Products: The

53

1270 AnaPurNa Study. J Nat Prod 75:1243–1255. doi: 10.1021/np300066q

1271 Pedersen MB, Gaudu P, Lechardeur D, et al (2012) Aerobic Respiration Metabolism in Lactic Acid Bacteria

1272 and Uses in Biotechnology. Annu Rev Food Sci Technol 3:37–58. doi: 10.1146/annurev-food-022811-

1273 101255

1274 Pei L, Schmidt M (2016) Sustainable Assessment on Using Bacterial Platform to Produce High-Added-

1275 Value Products from Berries through Metabolic Engineering. In: Singh S (ed) Systems Biology

1276 Application in Synthetic Biology. Springer India, pp 71–78

1277 Pontes DS, de Azevedo MSP, Chatel J-M, et al (2011) Lactococcus lactis as a live vector: Heterologous

1278 protein production and DNA delivery systems. Protein Expr Purif 79:165–175. doi:

1279 10.1016/j.pep.2011.06.005

1280 Prescott TAK, Ariño J, Kite GC, Simmonds MSJ (2012) Inhibition of human calcineurin and yeast

1281 calcineurin-dependent gene expression by Jasminum humile leaf and root extracts. J Ethnopharmacol

1282 140:293–297. doi: 10.1016/j.jep.2012.01.020

1283 Prince K, Smith M (2004) Protein Purification Protocols. In: Cutler P (ed) Protein Purification Protocols.

1284 Humana Press, Totowa, USA, pp 463–480

1285 Quideau S, Deffieux D, Douat-Casassus C, Pouységu L (2011) Plant Polyphenols: Chemical Properties,

1286 Biological Activities, and Synthesis. Angew Chemie Int Ed 50:586–621. doi: 10.1002/anie.201000044

1287 Rani MU, Appaiah A (2011) Optimization of culture conditions for bacterial cellulose production from

1288 Gluconacetobacter hansenii UAC09. Ann Microbiol 61:781–787. doi: 10.1007/s13213-011-0196-7

1289 Rasmussen SE, Frederiksen H, Struntze Krogholm K, Poulsen L (2005) Dietary proanthocyanidins:

1290 Occurrence, dietary intake, bioavailability, and protection against cardiovascular disease. Mol Nutr

1291 Food Res 49:159–174. doi: 10.1002/mnfr.200400082

54

1292 Ratnam BVV, Narasimha Rao M, Damodar Rao M, et al (2003) Optimization of fermentation conditions for

1293 the production of ethanol from sago starch using response surface methodology. World J Microbiol

1294 Biotechnol 19:523–526. doi: 10.1023/A:1025174731814

1295 Renaud S, de Lorgeril M (1992) Wine, alcohol, platelets, and the French paradox for coronary heart disease.

1296 Lancet 339:1523–1526. doi: 10.1016/0140-6736(92)91277-F

1297 Ro D-K, Paradise EM, Ouellet M, et al (2006) Production of the antimalarial drug precursor artemisinic acid

1298 in engineered yeast. Nature 440:940–943. doi: 10.1038/nature04640

1299 Ruiz A, Hermosı́n-Gutiérrez I, Mardones C, et al (2010) Polyphenols and Antioxidant Activity of Calafate

1300 (Berberis microphylla) Fruits and Other Native Berries from Southern Chile. J Agric Food Chem

1301 58:6081–6089. doi: 10.1021/jf100173x

1302 Rusnak F, Mertz P (2000) Calcineurin: form and function. Physiol Rev 80:1483–521.

1303 Santos CNS, Koffas M, Stephanopoulos G (2011) Optimization of a heterologous pathway for the

1304 production of flavonoids from glucose. Metab Eng 13:392–400. doi: 10.1016/j.ymben.2011.02.002

1305 SCBD (2011) Nagoya Protocol on access to genetic resources and the fair and equitable sharing of benefits

1306 arising from their utilization to the convention on biological diversity. Quebec, Canada

1307 Secretary of State (UK) (2015) Nagoya Protocol (Compliance) Regulations 2015.

1308 Segrè D, Vitkup D, Church GM (2002) Analysis of optimality in natural and perturbed metabolic networks.

1309 Proc Natl Acad Sci U S A 99:15112–7. doi: 10.1073/pnas.232349399

1310 Shen X-H, Zhou N-Y, Liu S-J (2012) Degradation and assimilation of aromatic compounds by

1311 Corynebacterium glutamicum: another potential for applications for this bacterium? Appl Microbiol

1312 Biotechnol 95:77–89. doi: 10.1007/s00253-012-4139-4

1313 Shen X, Liu S (2005) Key enzymes of the protocatechuate branch of the β-ketoadipate pathway for aromatic

55

1314 degradation in Corynebacterium glutamicum. Sci China Ser C Life Sci 48:241–249. doi:

1315 10.1007/BF03183617

1316 Shinfuku Y, Sorpitiporn N, Sono M, et al (2009) Development and experimental verification of a genome-

1317 scale metabolic model for Corynebacterium glutamicum. Microb Cell Fact 8:43. doi: 10.1186/1475-

1318 2859-8-43

1319 Shults CW (2006) Lewy bodies. Proc Natl Acad Sci U S A 103:1661–8. doi: 10.1073/pnas.0509567103

1320 Simeonidis E, Price ND (2015) Genome-scale modeling for metabolic engineering. J Ind Microbiol

1321 Biotechnol 42:327–338. doi: 10.1007/s10295-014-1576-3

1322 Song AA, Abdullah JO, Abdullah MP, et al (2014) Engineering the lactococcal mevalonate pathway for

1323 increased sesquiterpene production. FEMS Microbiol Lett 355:177–84. doi: 10.1111/1574-6968.12469

1324 Stahlhut SG, Siedler S, Malla S, et al (2015) Assembly of a novel biosynthetic pathway for production of the

1325 plant flavonoid fisetin in Escherichia coli. Metab Eng 31:84–93. doi: 10.1016/j.ymben.2015.07.002

1326 Stanford NJ, Millard P, Swainston N (2015) RobOKoD: microbial strain design for (over)production of

1327 target compounds. Front Cell Dev Biol 3:17. doi: 10.3389/fcell.2015.00017

1328 Stavang JA, Freitag S, Foito A, et al (2015) Raspberry fruit quality changes during ripening and storage as

1329 assessed by colour, sensory evaluation and chemical analyses. Sci Hortic (Amsterdam) 195:216–225.

1330 doi: 10.1016/j.scienta.2015.08.045

1331 Stracke R, Werber M, Weisshaar B (2001) The R2R3-MYB gene family in Arabidopsis thaliana. Curr Opin

1332 Plant Biol 4:447–456. doi: 10.1016/S1369-5266(00)00199-0

1333 Su LJ, Auluck PK, Outeiro TF, et al (2010) Compounds from an unbiased chemical screen reverse both ER-

1334 to-Golgi trafficking defects and mitochondrial dysfunction in Parkinson’s disease models. Dis Model

1335 Mech 3:194–208. doi: 10.1242/dmm.004267

56

1336 Suh Y, Afaq F, Johnson JJ, Mukhtar H (2008) A plant flavonoid fisetin induces apoptosis in colon cancer

1337 cells by inhibition of COX2 and Wnt/EGFR/NF- B-signaling pathways. Carcinogenesis 30:300–307.

1338 doi: 10.1093/carcin/bgn269

1339 Tardiff DF, Tucci ML, Caldwell KA, et al (2012) Different 8-hydroxyquinolines protect models of TDP-43

1340 protein, α-synuclein, and polyglutamine proteotoxicity through distinct mechanisms. J Biol Chem

1341 287:4107–20. doi: 10.1074/jbc.M111.308668

1342 Tayarani-Najaran Z, Mousavi SH, Tajfard F, et al (2013) Cytotoxic and apoptogenic properties of three

1343 isolated diterpenoids from Salvia chorassanica through bioassay-guided fractionation. Food Chem

1344 Toxicol 57:346–351. doi: 10.1016/j.fct.2013.03.037

1345 Tepper N, Shlomi T (2010) Predicting metabolic engineering knockout strategies for chemical production:

1346 accounting for competing pathways. Bioinformatics 26:536–43. doi: 10.1093/bioinformatics/btp704

1347 Tohge T, Nishiyama Y, Hirai MY, et al (2005) Functional genomics by integrated analysis of metabolome

1348 and transcriptome of Arabidopsis plants over-expressing an MYB transcription factor. Plant J 42:218–

1349 235. doi: 10.1111/j.1365-313X.2005.02371.x

1350 Tö rrö nen R (2009) Sources and health effects of dietary ellagitannins. In: Quideau S (ed) Chemistry and

1351 Biology of Ellagitannins: An Underestimated Class of Bioactive Plant Polyphenols. World Scientific

1352 Publishing, Singapore, p 298−319

1353 Vagiri M, Ekholm A, Öberg E, et al (2013) Phenols and ascorbic acid in black currants (Ribes nigrum L.):

1354 variation due to genotype, location, and year. J Agric Food Chem 61:9298–306. doi: 10.1021/jf402891s

1355 van Summeren-Wesenhagen P V, Marienhagen J (2015) Metabolic Engineering of Escherichia coli for the

1356 Synthesis of the Plant Polyphenol Pinosylvin. Appl Environ Microbiol 81:840–849. doi:

1357 10.1128/AEM.02966-14

1358 Vance C, Rogelj B, Hortobágyi T, et al (2009) Mutations in FUS, an RNA processing protein, cause familial

57

1359 amyotrophic lateral sclerosis type 6. Science 323:1208–11. doi: 10.1126/science.1165942

1360 Vazquez-Albacete D, Cavaleiro AM, Christensen U, et al (2016) An expression tag toolbox for microbial

1361 production of membrane bound plant cytochromes P450. Biotechnol Bioeng. doi: 10.1002/bit.26203

1362 Vera J, González-Alcón C, Marín-Sanguino A, Torres N (2010) Optimization of biochemical systems

1363 through mathematical programming: Methods and applications. Comput Oper Res 37:1427–1438. doi:

1364 10.1016/j.cor.2009.02.021

1365 Veríssimo A, Bassard J-E, Julien-Laferrière A, et al MassBlast: A workflow to accelerate RNA-seq and

1366 DNA database analysis.

1367 Vogt T (2010) Phenylpropanoid Biosynthesis. Mol Plant 3:2–20. doi: 10.1093/mp/ssp106

1368 Waldo GS, Standish BM, Berendzen J, Terwilliger TC (1999) Rapid protein-folding assay using green

1369 fluorescent protein. Nat Biotechnol 17:691–695. doi: 10.1038/10904

1370 Wang J, Guleria S, Koffas MA, Yan Y (2016a) Microbial production of value-added nutraceuticals. Curr

1371 Opin Biotechnol 37:97–104. doi: 10.1016/j.copbio.2015.11.003

1372 Wang Y, Bhuiya MW, Zhou R, Yu O (2015) Pterostilbene production by microorganisms expressing

1373 resveratrol O-methyltransferase. Ann Microbiol 65:817–826. doi: 10.1007/s13213-014-0922-z

1374 Wang Y, Zhu J, Meng X, et al (2016b) Comparison of polyphenol, anthocyanin and antioxidant capacity in

1375 four varieties of Lonicera caerulea berry extracts. Food Chem 197:522–529. doi:

1376 10.1016/j.foodchem.2015.11.006

1377 Watts KR, Ratnam J, Ang K-H, et al (2010) Assessing the trypanocidal potential of natural and semi-

1378 synthetic diketopiperazines from two deep water marine-derived fungi. Bioorg Med Chem 18:2566–

1379 2574. doi: 10.1016/j.bmc.2010.02.034

1380 Weckwerth W (2003) Metabolomics in systems biology. Annu Rev Plant Biol 54:669–689. doi:

58

1381 10.1146/annurev.arplant.54.031902.135014

1382 Weller MG, G. M (2012) A Unifying Review of Bioassay-Guided Fractionation, Effect-Directed Analysis

1383 and Related Techniques. Sensors 12:9181–9209. doi: 10.3390/s120709181

1384 Wen Z, Minton NP, Zhang Y, et al (2016) Enhanced solvent production by metabolic engineering of a twin-

1385 clostridial consortium. Metab Eng. doi: 10.1016/j.ymben.2016.10.013

1386 Xu P, Ranganathan S, Fowler ZL, et al (2011) Genome-scale metabolic network modeling results in minimal

1387 interventions that cooperatively force carbon flux towards malonyl-CoA. Metab Eng 13:578–587. doi:

1388 10.1016/j.ymben.2011.06.008

1389 Xu P, Wang W, Li L, et al (2013) Design and Kinetic Analysis of a Hybrid Promoter–Regulator System for

1390 Malonyl-CoA Sensing in Escherichia coli. doi: 10.1021/cb400623m

1391 Yang X, Summerhurst DK, Koval SF, et al (2001) Isolation of an antimicrobial compound from Impatiens

1392 balsamina L. using bioassay-guided fractionation. Phyther Res 15:676–680. doi: 10.1002/ptr.906

1393 Yu C, Cao Y, Zou H, Xian M (2011) Metabolic engineering of Escherichia coli for biotechnological

1394 production of high-value organic acids and alcohols. Appl Microbiol Biotechnol 89:573–583. doi:

1395 10.1007/s00253-010-2970-z

1396 Zadernowski R, Naczk M, Nesterowicz J (2005) Phenolic Acid Profiles in Some Small Berries. J Agric Food

1397 Chem 53:2118–2124. doi: 10.1021/JF040411P

1398 Zelle RM, de Hulster E, Kloezen W, et al (2010) Key process conditions for production of C(4) dicarboxylic

1399 acids in bioreactor batch cultures of an engineered Saccharomyces cerevisiae strain. Appl Environ

1400 Microbiol 76:744–50. doi: 10.1128/AEM.02396-09

1401 Zhang H, Matsuda H, Yamashita C, et al (2009) from the processed leaves of Hydrangea

1402 macrophylla var. thunbergii as a new type of anti-diabetic compound. Eur J Pharmacol 606:255–261.

59

1403 doi: 10.1016/j.ejphar.2009.01.005

1404 Zheng J, Ding C, Wang L, et al (2011) Anthocyanins composition and antioxidant activity of wild Lycium

1405 ruthenicum Murr. from Qinghai-Tibet Plateau. Food Chem 126:859–865. doi:

1406 10.1016/j.foodchem.2010.11.052

1407 Zomorrodi AR, Suthers PF, Ranganathan S, Maranas CD (2012) Mathematical optimization applications in

1408 metabolic networks. Metab Eng 14:672–686. doi: 10.1016/j.ymben.2012.09.005

1409

1410

60

1411 Figure legends

1412 Figure 1: a) The molecular structure of the stilbene trans-resveratrol. b) Basic flavonoid structure. Rx are

1413 positions where decorating groups (e.g. hydroxyl, acyl, glycosyl, etc.), which differentiate the compounds

1414 and alter their chemical properties, could be attached.

1415 Figure 2: Principal component analysis (PCA) plots of the full dataset without reference material and

1416 blanks. Each dot represents the profile of one sample aliquot whereas lines delimit groups of samples that

1417 cluster together. a) PCA plots of the full dataset without reference material and blanks in positive mode.

1418 Components (Scr) 1 and 2 explain 9.79% and 7.59% of the variation respectively. b) PCA plots of the full

1419 dataset without reference material and blanks in negative mode. Components (Scr) 1 and 2 explain 13.20%

1420 and 10.30% of the variation respectively.

1421 Figure 3: Schematic representation of yeast models. a. Yeast models of neurodegenerative diseases.

1422 Humanized yeast strains encoding chimeric fusions of disease genes with GFP (green fluorescent protein)

1423 under the control of a galactose-inducible (GAL1) promoter. Expression of Aβ42, αSyn (SNCA gene), FUS

1424 and HTTpQ103 leads to the formation of toxic aggregates, recapitulating the pathological processes of

1425 Alzheimer’s disease (AD), Parkinson’s disease (PD), Amyotrophic lateral sclerosis (ALS), and Huntington’s

1426 disease (HD), respectively. b. Yeast model of Crz1 (NFAT orthologue) activation. Crz1 is activated by the

1427 serine/threonine protein phosphatase calcineurin (composed by CnA and CnB subunits). The reporter strain

1428 encodes the lacZ gene under the control of a promoter containing Crz1-binding sites, allowing the

1429 assessment of Crz1 activation through the measurement of β-galactosidase activity.

1430 Figure 4: Identification of bioactivities in the berry germplasm. a) Strategy used for the discovery of

1431 bioactive compounds for Alzheimer’s disease. BY4741_erg6 recombinant yeast cells expressing GFP-Aβ42

1432 were grown in SD galactose medium and cell viability was assessed by growth curves for 24 h in cells

1433 challenged or unchallenged with Lycium chinense polyphenol extracts. Cells containing the empty vector

1434 were used as controls. The areas under the curve (AUC) were integrated using the Origin software

61

1435 (OriginLab, Northampton, MA). Values represent the mean ± standard error of mean (SEM) of at least three

1436 biological replicates.

1437 b) Strategy used for the discovery of bioactive compounds with anti-inflammatory potential. BY4741 yeast

1438 cells encoding CDREpromoter-lacZ were treated or not with Lycium chinense polyphenol extracts and induced

1439 with 1.8 mM MnCl2. Crz1 activation was assessed by monitoring β-galactosidase activity using ortho-

1440 nitrophenyl-β-galactoside (ONPG). The immunosuppressant FK506, a well-known inhibitor of calcineurin

1441 (Dumont 2000) was used as a positive control. The values represent the mean ± SEM of at least three

1442 biological replicates, *p < 0.05, **p < 0.01, ***p < 0.001.

1443 Figure 5. Yeast as a discovery platform of therapeutic compounds. Iterative bio-guided fractionation of

1444 complex mixtures allows the discovery of small molecules improving cell growth, by rescue of protein

1445 aggregates toxicity, or inhibiting inflammatory processes. High-throughput formats aligned with chemical

1446 identification by LC-MS approaches accelerate the identification of candidate compounds.

1447 Figure 6: Polyphenol biosynthetic pathway in plants (modified from Falcone Ferreyra et al. 2012). PAL,

1448 phenylalanine ammonia-lyase; C4H, cinnamate 4-hydroxylase; 4CL, 4-coumaroyl-CoA ligase; CHS,

1449 chalcone synthase; CHI, chalcone isomerase; F3H, flavanone 3-hydroxylase; F3’5’H, flavonoid 3’-5’-

1450 hydroxylase; DFR, dihydroflavonol 4-reductase; ANS, anthocyanidin synthase; ANR, anthocyanidin

1451 reductase; UFGT, flavonoid 3-O-glucosyltransferase. LAR, leucoanthocyanidin reductase; FNS, flavone

1452 synthase; FLS, flavonol synthase; STS, stilbene synthase. P450 enzymes are highlighted in red.

1453 Figure 7: Overview of the endogenous phenylpropanoid degradation and the engineered pathway for

1454 polyphenol synthesis in C. glutamicum. One of the precursors for the production of the stilbene trans-

1455 resveratrol (catalyzed by stilbene synthase, STS) or for naringenin chalcone (catalyzed by chalcone

1456 synthases, CHS) is p-coumaroyl-CoA. Naringenin chalcone isomerizes to the (2S)-flavanone naringenin

1457 either spontaneously or catalyzed by the activity of chalcone isomerase (CHI). In C. glutamicum, p-coumaric

1458 acid can be degraded to 4-hydroxybenzoate by a CoA-dependent, β-oxidative deacetylation pathway. 4-

1459 Hydroxybenzoate is subsequently hydroxylated to protocatechuate, which is catabolized to succinyl-CoA

62

1460 and acetyl-CoA by the β-ketoadipate pathway. Genes coding for the underlined enzymes were deleted in the

1461 course of the construction of the platform strain C. glutamicum DelAro4.

1462 Figure 8: Heterologous biosynthetic pathway for fisetin assembled in E. coli. TAL, tyrosine ammonia-lyase;

1463 4CL, 4-coumaroyl-CoA ligase; CHS::CHR, chalcone synthase::chalcone reductase fusion; CHI, chalcone

1464 isomerase; F3H, flavanone 3-hydroxylase; FLS, flavonol synthase; F3’H::CPR, flavonoid 3’-

1465 hydroxylase::cytochrome P450 reductase fusion.

1466 Figure 9: Heterologous biosynthetic pathways introduced into the genome-scale models of C. glutamicum

1467 and L. lactis leading to the four target compounds: i) trans-resveratrol ii) pelargonidin iii) quercetin and iv)

1468 fisetin. Two heterologous enzymes (TAL and 4CL) are common for the biosynthesis of the phenolic

1469 molecules. Enzyme abbreviations: TAL, tyrosine ammonia-lyase; 4CL, 4-coumaroyl-CoA ligase; STS,

1470 stilbene synthase; CHS, chalcone synthase; CHR, chalcone reductase; CHI, chalcone isomerase; F3H,

1471 flavanone 3–hydroxylase; FLS, flavonol synthase; F3’H, flavonoid 3' hydroxylase; CPR, cytochrome P450

1472 reductase.

1473 Figure 10: Partition coefficients in the form of log P values of trans-resveratrol using different organic

1474 solvents using C. glutamicum fermentation broth as the aqueous phase.

1475 Figure 11: On the left, protein concentration (expressed as percentage of the initial broth protein

1476 concentration) in the organic and aqueous phases, after performing liquid extraction. The concentration in

1477 the organic phase is almost zero (considering the error bars), indicating that some protein stayed in the

1478 interphase. On the right, the formation of an interphase after the broth had been extracted with organic

1479 solvent (a mixture of heptanoic acid and hexyl acetate in this case) was clear.

1480 Figure 12: Chromatogram following a pulse injection of 10 ml of C. glutamicum spent fermentation broth in

1481 a 30 ml column packed with Amberlite XAD-16 resin. A washing step was performed with 10 column

1482 volumes (CVs) of MilliQ water. Afterwards, elution was performed with a water/ethanol gradient for 12 CV,

1483 followed by a final regeneration step with 100% ethanol.

63

1484 Figure 13: Experimental set-up for the “Continuous Product Removal” mode.

1485

64

1486 Tables

1487 Table 1: Anthocyanidin structures (aglycone form) found in different berry species. The presence of a

1488 compound is noted with + whereas the absence is noted with -.

Type Pelargonidin Cyanidin Delphinidin Peonidin Petunidin Malvinidin

Structure

Molecular + + + + + + C15H11O5 C15H11O6 C15H11O7 C16H13O6 C16H13O7 C17H15O7 formula

Fragaria1 + + - - - -

Ribes1 + + + - + -

Rubus1 + + - - - -

Vaccinium1 - + + + + +

Vitis1 - + + + + +

Lonicera2,5 + + + + - +

Lycium3 - - + - + +

Berberis4 - + + + + +

Ugni4 - + - + - -

Aristotelia6 - + + - - -

1489 1 Phenol explorer database (http://phenol-explorer.eu); 2 (Chaovanalikit et al. 2004); 3 (Zheng et al. 2011); 4

1490 (Ruiz et al. 2010); 5 (Wang et al. 2016b); 6 (Fredes et al. 2014)

1491

1492

65

1493 Table 2: Selected species for bioactivity testing according to the phytochemical diversity.

Species Variety/line Species Variety/line

Aristotelia chilensis - Rubus armeniacus Focke Himalayan Giant (Molina)Stuntz

Berberis buxfolia Lam. - Rubus occidentalis L. Huron

Lonicera caerulea L. S322-23 Rubus loganobaccus L.H.Bailey Sunberry

Lycium chinense Mill. - Rubus loganobaccus L.H.Bailey Tayberry

Ribes grossularia L. 9294 Rubus bartonii Newton Ashton Cross

Ribes grossularoides Maxim. H1-12-1 Rubus fruticosus L. agg. Fantasia

Ribes nevadense Kellog - Rubus brigantinus Samp. -

Ribes spp. 644217 Rubus genevierii Boreau. -

Ribes spp. 1126 Rubus henriquesii Samp. -

Ribes spp. 2292-1 Rubus hochstetterorum Seub. -

Rubus sampaioanus Sudre ex Ribes spp. Muravushka - Samp.

Ribes spp. - Rubus vagabundus Samp. -

Ribes petraeum Wulf. R2-1-2 Rubus idaeus L. Octavia

Ribes x saundersii Jancz. XB4 Rubus idaeus L. Prestige

Ribes bethmontii Jancz. XW6 Ugni molinae Turcz. Red Pearl

Ribes rubrum L. S11-3-36 Vaccinium uliginosum L. -

Rubus spp. B14 Vaccinium vitis-idaea L. -

1494

1495

66

1496 Table 3: Transcriptome resources for berries developed on BacHBerry

Species Plant material Total number of raw reads

Aristotelia sp. ripe berries 397,707,372

Berberis sp. ripe berries 444,362,698

Corema sp. leaf 353,604,932

Lonicera sp. ripe berries 397,214,254

Ribes sp.1 ripe berries 336,479,242

Ribes sp.2 ripe berries 393,665,630

Rubus sp.1 berries (3 stages) 1,040,224,680

Rubus sp.2 berries (3 stages) 1,064,858,518

Rubus sp.3 ripe berries 505,754,030

Rubus sp.4 ripe berries 390,608,452

Ugni sp. ripe berries 405,024,920

Vaccinium sp.1 ripe berries 373,159,882

Vaccinium sp.2 ripe berries 375,778,718

1497

1498

67

1499 Table 4: Statistics on five major enzyme families representing conserved biochemical steps of the

1500 phenylpropanoid pathway. Candidates displayed more than 70% sequence coverage to reference query

1501 sequences. This work led to the creation of a large dataset of candidates used for pathway elucidation and

1502 reconstitution in bacterial hosts.

Enzyme families Candidates per transcriptome (min-max)

Conserved phenylpropanoid pathway enzymes 109-180

CYPs 100-177

UGTs 70-145

2-OGDs 32-88

O-methyltransferases 8-51

Acyltransferases 23-36

1503

1504

68

1505 Table 5: Summary of the outcomes of the project

Specific Bioprospecting for discovery of Characterization of phenolics Bacterial cell factories for Fermentation and application novel berry phenolics biosynthetic pathways production of berry phenolics bioprocess engineering area in project General aims To tap into the biodiversity of berries Identification and characterization Design and generation of bacterial Implementation of production of high- of BacHBerry from around the globe and of their of berry phenolics biosynthetic cell factories for production of high- value phenolics in fermenters up to phenolics content pathways value berry phenolics demonstration scale Achievements • Standardization of methodologies • Transcriptomes of 13 germplasms • L. lactis strains were engineered for • Cultivation conditions for improved of the project for harvesting, extract preparation, spanning eight genera, seven stilbene (trans-resveratrol and productivity were developed for both phenolics content assessment and families and seven orders were pterostilbene) and anthocyanin L. lactis and C. glutamicum through fractionation generated and analyzed production optimization of fermentation parameters (batch versus fed-batch • Metabolomics data from berry •New algorithms for functional • Production of various stilbenes operation, substrate and dissolved species originating from UK, genomics and improved (trans-resveratrol, pinosylvin, and oxygen concentration, etc.) mainland Europe, Russia, Chile, and computational methods for piceatannol), methylated stilbenes China was obtained and made pathway identification were (pinostilbene and pterostilbene), • Whey was evaluated as an alternative available in form of an online established flavanones (naringenin, pinocembrin, carbon source for polyphenol database and eryodictiol), and flavonols production in L lactis • Over 4000 candidate genes for (kaempferol and quercetin) was • Implementation of robust assays various biosynthetic steps, achieved in C. glutamicum with • Multiple bioseparation methods for based on the SMART platform for transport and regulation of yields comparable to those obtained extraction of produced polyphenols discovery of bioactivity and polyphenol production were in the model organism E. coli. from fermentation broth were functionality identified Production titers for the produced evaluated

69

flavonols are the highest titers • Obtained berry extracts were • Biochemical activities of some of obtained in engineered • Two possible designs for a system for assayed for bioactivity against the gene candidates were tested, microorganism until today continuous product removal from the several human diseases, and thus validating the predictions fermentation broth were implemented additionally evaluated for other • Polyphenol production was further functional uses (eq. food additives, improved by engineering enhanced • Resveratrol production in C.

antimicrobials). Bioactivities against precursor supply (eq. L-tyrosine, glutamicum was scaled up from shake Alzheimer’s, Parkinson’s and malonyl-CoA) via rational design, flask to 5 l fermenter to demonstration Huntington’s diseases, Amyotrophic modeling-based prediction, and use scale (250 l) Lateral Sclerosis and inflammation of biosensors were detected. • Alternative biosynthetic routes were • Several pure bioactive effector explored though engineering of compounds were identified and trans-resveratrol production from 4- validated hydroxybenzoic acid (HBA) in C. glutamicum

• Computational models were developed allowing predicting a minimal set of modifications needed for improved polyphenol production in both a single-strain population and a consortium of two or more species 1506

70

1507

71

Figure 1: a) The molecular structure of the stilbene trans-resveratrol. b) Basic flavonoid structure. Rx are positions where decorating groups (e.g. hydroxyl, acyl, glycosyl, etc.), which differentiate the compounds and alter their chemical properties, could be attached.

Figure 2: Principal component analysis (PCA) plots of the full dataset without reference material and blanks. Each dot represents the profile of one sample aliquot whereas lines delimit groups of samples that cluster together. a) PCA plots of the full dataset without reference material and blanks in positive mode. Components (Scr) 1 and 2 explain 9.79% and 7.59% of the variation respectively. b) PCA plots of the full dataset without reference material and blanks in negative mode. Components (Scr) 1 and 2 explain 13.20% and 10.30% of the variation respectively.

Figure 3: Schematic representation of yeast models. a. Yeast models of neurodegenerative diseases. Humanized yeast strains encoding chimeric fusions of disease genes with GFP (green fluorescent protein) under the control of a galactose-inducible (GAL1) promoter. Expression of Aβ42, αSyn (SNCA gene), FUS and HTTpQ103 leads to the formation of toxic aggregates, recapitulating the pathological processes of Alzheimer’s disease (AD), Parkinson’s disease (PD), Amyotrophic lateral sclerosis (ALS), and Huntington’s disease (HD), respectively. b. Yeast model of Crz1 (NFAT orthologue) activation. Crz1 is activated by the serine/threonine protein phosphatase calcineurin (composed by CnA and CnB subunits). The reporter strain encodes the lacZ gene under the control of a promoter containing Crz1-binding sites, allowing the assessment of Crz1 activation through the measurement of β-galactosidase activity.

Figure 4: Identification of bioactivities in the berry germplasm. a) Strategy used for the discovery of bioactive compounds for Alzheimer’s disease. BY4741_erg6 recombinant yeast cells expressing GFP-Aβ42 were grown in SD galactose medium and cell viability was assessed by growth curves for 24 h in cells challenged or unchallenged with Lycium chinense polyphenol extracts. Cells containing the empty vector were used as controls. The areas under the curve (AUC) were integrated using the Origin software (OriginLab, Northampton, MA). Values represent the mean ± standard error of mean (SEM) of at least three biological replicates. b) Strategy used for the discovery of bioactive compounds with anti-inflammatory potential. BY4741 yeast cells encoding CDREpromoter-lacZ were treated or not with Lycium chinense polyphenol extracts and induced with 1.8 mM MnCl2. Crz1 activation was assessed by monitoring β-galactosidase activity using ortho- nitrophenyl-β-galactoside (ONPG). The immunosuppressant FK506, a well-known inhibitor of calcineurin (Dumont 2000) was used as a positive control. The values represent the mean ± SEM of at least three biological replicates, *p < 0.05, **p < 0.01, ***p < 0.001.

Figure 5. Yeast as a discovery platform of therapeutic compounds. Iterative bio-guided fractionation of complex mixtures allows the discovery of small molecules improving cell growth, by rescue of protein aggregates toxicity, or inhibiting inflammatory processes. High-throughput formats aligned with chemical identification by LC-MS approaches accelerate the identification of candidate compounds.

Figure 6: Polyphenol biosynthetic pathway in plants (modified from Falcone Ferreyra et al. 2012). PAL, phenylalanine ammonia-lyase; C4H, cinnamate 4-hydroxylase; 4CL, 4-coumaroyl-CoA ligase; CHS, chalcone synthase; CHI, chalcone isomerase; F3H, flavanone 3-hydroxylase; F3’5’H, flavonoid 3’-5’- hydroxylase; DFR, dihydroflavonol 4-reductase; ANS, anthocyanidin synthase; ANR, anthocyanidin reductase; UFGT, flavonoid 3-O-glucosyltransferase. LAR, leucoanthocyanidin reductase; FNS, flavone synthase; FLS, flavonol synthase; STS, stilbene synthase. P450 enzymes are highlighted.

protocatechuate 3-carboxy-cis,cis-muconate O O O - tricarboxylic - PcaGH HO HO O O acid cycle glucose HO HO O shikimate PcaD pathway PobA PcaC succinyl-CoA PcaB - O O O PcaIJ O - SCoA - O O PcaF OH O - O O O HO NH O + 2 L-tyrosine HO SCoA O 3-oxoadipate TAL PhdE 4-hydroxybenzoate acetyl-CoA PhdB O PhdC OH acid HO p-coumaric O CHS O OH 4CL SCoA HO + 3 malonyl-CoA p-coumaroyl-CoA HO HO OH naringenin CHI STS + 3 malonyl-CoA chalcone

O OH OH

O OH

OH HO HO resveratrol naringenin

Figure 7: Overview of the endogenous phenylpropanoid degradation and the engineered pathway for polyphenol synthesis in C. glutamicum. One of the precursors for the production of the stilbene trans- resveratrol (catalyzed by stilbene synthase, STS) or for naringenin chalcone (catalyzed by chalcone synthases, CHS) is p-coumaroyl-CoA. Naringenin chalcone isomerizes to the (2S)-flavanone naringenin either spontaneously or catalyzed by the activity of chalcone isomerase (CHI). In C. glutamicum, p-coumaric acid can be degraded to 4-hydroxybenzoate by a CoA-dependent, β-oxidative deacetylation pathway. 4- Hydroxybenzoate is subsequently hydroxylated to protocatechuate, which is catabolized to succinyl-CoA and acetyl-CoA by the β-ketoadipate pathway. Genes coding for the underlined enzymes were deleted in the course of the construction of the platform strain C. glutamicum DelAro4.

Figure 8: Heterologous biosynthetic pathway for fisetin assembled in E. coli. TAL, tyrosine ammonia-lyase; 4CL, 4-coumaroyl-CoA ligase; CHS::CHR, chalcone synthase::chalcone reductase fusion; CHI, chalcone isomerase; F3H, flavanone 3-hydroxylase; FLS, flavonol synthase; F3’H::CPR, flavonoid 3’- hydroxylase::cytochrome P450 reductase fusion.

Figure 9: Heterologous biosynthetic pathways introduced into the genome-scale models of C. glutamicum and L. lactis leading to the four target compounds: i) trans-resveratrol ii) pelargonidin iii) quercetin and iv) fisetin. Two heterologous enzymes (TAL and 4CL) are common for the biosynthesis of the phenolic molecules. Enzyme abbreviations: TAL, tyrosine ammonia-lyase; 4CL, 4-coumaroyl-CoA ligase; STS, stilbene synthase; CHS, chalcone synthase; CHR, chalcone reductase; CHI, chalcone isomerase; F3H, flavanone 3–hydroxylase; FLS, flavonol synthase; F3’H, flavonoid 3' hydroxylase; CPR, cytochrome P450 reductase.

Figure 10: Partition coefficients in the form of log P values of trans-resveratrol using different organic solvents using C. glutamicum fermentation broth as the aqueous phase.

Figure 11: On the left, protein concentration (expressed as percentage of the initial broth protein concentration) in the organic and aqueous phases, after performing liquid extraction. The concentration in the organic phase is almost zero (considering the error bars), indicating that some protein stayed in the interphase. On the right, the formation of an interphase after the broth had been extracted with organic solvent (a mixture of heptanoic acid and hexyl acetate in this case) was clear.

Figure 12: Chromatogram following a pulse injection of 10 ml of C. glutamicum spent fermentation broth in a 30 ml column packed with Amberlite XAD-16 resin. A washing step was performed with 10 column volumes (CVs) of MilliQ water. Afterwards, elution was performed with a water/ethanol gradient for 12 CV, followed by a final regeneration step with 100% ethanol.

Figure 13: Experimental set-up for the “Continuous Product Removal” mode.

SUPPLEMENTARY TABLES

Title

BacHBerry: BACterial Hosts for production of Bioactive phenolics from bERRY fruits

Running Title

BacHBerry

Authors

BacHBerry consortium

*Corresponding author:

Alexey Dudnik

Email: [email protected]

Phone: + 45 93 51 11 01

Fax: +45 45 25 80 01

Table S1 – Decorating glycosyl groups of anthocyanins found in different berry species. The presence of a glycosylation pattern is noted with + whereas the absence is noted with -.

- - - O - 5

- O -

- O 5

-

- 5

- rutinoside

- rutinoside

arabinoside glucoside galactoside rutinoside sambubioside sophoroside glucosyl sambubiosyl galactoside gentibioside rutinoside diglucoside ------xyloside - - - xylosyl - - - - - O - O - O - O - O - O - O - O - O - O - O - O - O 3 3 3 3 3 3 3 3,5 3 3 glucoside 3 3 glucoside 3 3 glucoside Fragaria1 + + - + ------Ribes1 - + + + + + - - + - + - - - Rubus1 - + - + - + + + + - + - - - Vaccinium1 + + + ------Vitis1 - + ------Lonicera2 - + - + - - - + - - - - + - Lycium3 ------+ - - - + - + Berberis4,5 - + - + + - - + - + - - - + Ugni4 - + ------1 Phenol explorer database (http://phenol-explorer.eu) 2 Chaovanalikit et al., 2004 3 Zheng et al., 2011 4 Ruiz et al., 2010 5 Ruiz et al., 2013

Table S2 – Decorating acyl groups of anthocyanins found in in a variety of berry species

- - - glycoside) glycoside) - - - glycoside) - glycoside)

- coumaryl

(6’’acetyl (6’’malonyl (6’’succinnyl (6’’dioxyalyl (feruloyl (6’’caffeoyl - - - - - (6’’p - -

- O - O - O - O - O - O - O 3 3 3 glycoside) 3 glycoside) 3 glycoside) 3 3

1 Fragaria - + + - - - - 1 Ribes - - - - + - + 1 Rubus - + - + - - - 1 Vaccinium + ------Vitis1,2 + - - - + - + Lycium3 - - - - + + +

1 Phenol explorer database (http://phenol-explorer.eu) 2 Figueiredo-Gonzalez et al. 2012 3 Zheng et al. 2011

Table S3 – List of all the species/varieties and harvest locations of the germplasm collections

Species Variety/line Harvest location Aristotelia chilensis (Molina)Stuntz - Chilei Berberis buxfolia Lam. - Chileii Lonicera caerulea L. 260-11 Russiaiii Lonicera caerulea L. 838-12 Russia Lonicera caerulea L. Avacha Russiaiii Lonicera caerulea L. Bazhovskaya Russiaiii Lonicera caerulea L. Leningradskii Velikan Russiaiii Lonicera caerulea L. Malvina Russiaiii Lonicera caerulea L. Morena Russiaiii Lonicera caerulea L. Nimfa Russiaiii Lonicera caerulea L. Pavlovskaya Russiaiii Lonicera caerulea L. S322-23 Russiaiii Lonicera caerulea L. Solovey Russiaiii Lonicera caerulea L. Start Russiaiii Lonicera caerulea L. Suvenir Russiaiii Lycium chinense Mill. - Chinaiv Ribes spp. L. 1126 United Kingdomv Ribes spp. L. 1196 United Kingdomv Ribes spp. L. 2174 United Kingdomv Ribes spp. L. 2292-1 United Kingdomv Ribes spp. L. 644217 United Kingdomv Ribes spp. L. - United Kingdomv Ribes spp. L. - Chileii Ribes aureum Pursh R2-1-2 United Kingdomv Ribes bethmontii Jancz. XW6 United Kingdomv Ribes distans Jancz. 2269 United Kingdomv Ribes divaricatum Dougl. Montana United Kingdomv Ribes fuscescens Jancz. XB6 United Kingdomv Ribes futurum Jancz. XR4 United Kingdomv Ribes giraldii Jancz. B2-3-1 United Kingdomv Ribes grossularia L. 1815-21 United Kingdomv Ribes grossularia L. 1824-37 United Kingdomv Ribes grossularia L. 9492-5 United Kingdomv Ribes grossularia L. 96101-1 United Kingdomv Ribes grossularoides Maxim. H1-12-1 United Kingdomv Ribes holosericeum Otto& A.Dietr. XR2 United Kingdomv Ribes irriguum Douglas H1-5-1 United Kingdomv Ribes koehneanum Jancz. XR3 United Kingdomv Ribes nevadense Kellog - United Kingdomv Ribes nigrum L. Ben Alder United Kingdomv Ribes nigrum subsp. europaeum х R. nigrum Almiai Russiaiii subsp. sibiricum x R.dikuscha Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.dikuscha x R.nigrum Azhurnaya Russiaiii subsp. scandinavicum Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.dikuscha x R.nigrum Bolero Russiaiii subsp. scandinavicum Ribes nigrum subsp. europaeum х R. nigrum Buraya Dal’nevostochnaya Russiaiii subsp. sibiricum Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.dikuscha x R.nigrum Charovnitsa Russiaiii subsp. scandinavicum Ribes nigrum subsp. europaeum х R. nigrum Dochka Russiaiii subsp. sibiricum x R.dikuscha Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.dikuscha x R.nigrum Krasa L’vova Russiaiii subsp. scandinavicum R.nigrum subsp. scandinavicum x R. Lazur’ Russiaiii dikuscha Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.dikuscha x R.nigrum Malen’kii Prints Russiaiii subsp. scandinavicum Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.dikuscha x R.nigrum Mila Russiaiii subsp. scandinavicum Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.dikuscha x R.nigrum Monisto Russiaiii subsp. scandinavicum Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.dikuscha x R.nigrum Muravushka Russiaiii subsp. scandinavicum x R.janczewskii Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.dikuscha x R.nigrum Ocharovanie Russiaiii subsp. scandinavicum Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.dikuscha x R.nigrum Orlovskaya serenada Russiaiii subsp. scandinavicum Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.dikuscha x R.nigrum Orlovskii Val’s Russiaiii subsp. scandinavicum Ribes nigrum subsp. europaeum х R. nigrum Sensei Russiaiii subsp. sibiricum x R.dikuscha Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.dikuscha x R.nigrum Sharovidnaya Russiaiii subsp. scandinavicum Ribes nigrum subsp. europaeum х R. nigrum Syuita Kievskaya Russiaiii subsp. sibiricum x R.dikuscha Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.nigrum subsp. Ukrainka Russiaiii scandinavicum Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.dikuscha x R.nigrum Veloi Russiaiii subsp. scandinavicum Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.dikuscha x R.nigrum Vospominanie Russiaiii subsp. scandinavicum Ribes nigrum subsp. europaeum х R. nigrum subsp. sibiricum x R.dikuscha x R.nigrum Zelenaya Dymka Russiaiii subsp. scandinavicum Ribes nigrum subsp. europaeum х R. nigrum Zhuravushka Russiaiii subsp. sibiricum x R.dikuscha Ribes petraeum Wulf. R2-1-2 United Kingdomv Ribes rotundifolium Michx. H1-3-1 United Kingdomv Ribes rubrum L. Red Poll United Kingdomv Ribes rubrum L. 1518 United Kingdomv Ribes rubrum L. 1169 United Kingdomv Ribes rubrum L. 1196-2 United Kingdomv Ribes rubrum L. 1757-2 United Kingdomv Ribes rubrum L. Red Wing United Kingdomv Ribes rubrum L. S11-3-36 United Kingdomv Ribes sativum Syme R7-3-3 United Kingdomv Ribes x saundersii Jancz. XB4 United Kingdomv Ribes succirubrum Zabel ex Jancz. XW8 United Kingdomv Ribes warcewiczii Jancz. R7-3-3 United Kingdomv Ribes watsonianum Koehne - United Kingdomv Rubus armeniacus Focke [= R. procerus Himalayan Giant United Kingdomv auct.]) Rubus bartonii Newton syn. ‘Ashton Cross’ Ashton Cross United Kingdomv Rubus fruticosus L. agg. B13 United Kingdomv Rubus fruticosus L. agg. B14 United Kingdomv Rubus fruticosus L. agg. Fantasia United Kingdomv Rubus fruticosus L. agg. Karaka Black United Kingdomv Rubus geoides Sm - Chileii Rubus ursinus x R. armeniacus x R. idaeus Black Butte United Kingdomv R. ursinus x R.ursinus x R.idaeus x R. Kotata United Kingdomv ursinus x R. allegheniensis x R. argutus Rubus ursinus x R.ursinus x R.idaeus Ollalie United Kingdomv Rubus idaeus L. Glen Ample United Kingdomv Rubus idaeus L. Glen Fyne United Kingdomv Rubus idaeus L. Malling Admiral United Kingdomv Rubus idaeus L. Malmer Szedler United Kingdomv Rubus idaeus L. Octavia United Kingdomv Rubus idaeus L. Polana United Kingdomv Rubus idaeus L. Prestige United Kingdomv Rubus idaeus L. Tulameen United Kingdomv Rubus loganobaccus L.H.Bailey Nectarberry United Kingdomv Rubus loganobaccus L.H.Bailey Boysenberry United Kingdomv Rubus loganobaccus L.H.Bailey Loganberry United Kingdomv Rubus loganobaccus L.H.Bailey LY59-10 United Kingdomv Rubus loganobaccus L.H.Bailey Riwaka Choice United Kingdomv Rubus loganobaccus L.H.Bailey Sunberry United Kingdomv Rubus loganobaccus L.H.Bailey Tayberry United Kingdomv Rubus occidentalis L. Black Hawk United Kingdomv Rubus occidentalis L. Huron United Kingdomv Rubus occidentalis L. Plum Farmer United Kingdomv Rubus phoenicolasius Maxim. Wineberry Portugalvi Rubus brigantinus Samp. - Portugalvi Rubus genevieri Boreau. - Portugalvi Rubus henriquesii Samp. - Portugalvi Rubus hochstetterorum Seub. - Portugalvi Rubus sampaioanus Sudre ex Samp. - Portugalvi Rubus vagabundus Samp. - Portugalvi Ugni molinae Turcz. Red Pearl Chilevii Vaccinium angustifolium Ait. ex Benth Blomidon Chinaviii Vaccinium corymbosum L. Bluecrop Chinaix Vaccinium darrowii Camp Misty Chinaix Vaccinium uliginosum L. - Chinaviii Vaccinium virgatum Ait. Climax Chinaix Vaccinium vitis-idaea L. - Chinaviii

i Sampling location: Coyhaique, Región de Aysén, Chile ii Sampling location: Punta Arenas, Región de Magallanes, Chile iii Vavilov Institute collection: Pavlovsk, St-Petersburg, Russia iv Sampling location: Beijing botanical garden, Beijing, China v The James Hutton Institute collection: Invergowrie, Scotland, UK vi Propagation site: Fataca Experimental Field, Odemira, Portugal vii Sampling location:Mataquito, Región del Maule, Chile viii Sampling location: Tahe, Heilongjiang Province, China ix Sampling location: Dalian, Liaoning Province, China SUPPLEMENTARY MATERIALS

Title

BacHBerry: BACterial Hosts for production of Bioactive phenolics from bERRY fruits

Running Title

BacHBerry

Authors

BacHBerry consortium

*Corresponding author:

Alexey Dudnik

Email: [email protected]

Phone: + 45 93 51 11 01

Fax: +45 45 25 80 01

S1: Materials and Methods

Plant material and extraction procedure: A total of 112 different cultivars and species from the Aristotelia, Berberis, Lonicera, Lycium, Ribes, Rubus, Ugni and Vaccinium genera grown in different locations (see Supplementary Table 1) were harvested manually in the field and kept under cool conditions until they were transferred to -20ºC storage. Frozen fruit of each species/cultivar was weighed (approximately 50 g) into a solvent-proof blender containing 150 ml of pre-cooled 50ng ml-1 Morin (Sigma-Aldrich, Gillingham, UK) solution prepared with 0.2% formic acid in methanol solution. Samples were then homogenized and subsequently filtered using Whatman filter paper grade 1. The resulting filtrate was aliquoted and solvent-dried using a speed-vac followed by lyophilization.

Dried extracts were flushed with N2 and stored at -20ºC until analysis by LC-ToF-MS.

Phenolic profile determination by LC-ToF-MS: Dried extracts from each species/cultivar were resolubilized in triplicate using 2 ml of a 75% methanol solution with 0.1% formic acid. From these extracts, 500 µl were decanted into filter vials, sealed with 0.45 m PTFE-lined screwcap (Thomson Instrument Company, London, UK) and transferred into the autosampler. The analysis was achieved in positive and negative modes across 9 batches each, using an Agilent LC-ToF-MS system (Agilent Technologies, Cheadle, Cheshire, UK) consisting of a quaternary pump (Agilent 1260), a diode-array- detector (DAD) (Agilent 1260) a temperature control device (Agilent 1260) and a Thermostat (Agilent 1290) coupled to an Agilent 6224 time-of-flight (ToF) instrument. Five microliters of the sample were injected onto a 2x150 mm (4 µm) C18 column fitted with a C18 4 x 2 mm Security Guard™ cartridge (Phenomenex, Torrance, CA, USA). Sample and column temperatures were maintained at 4ºC and 30ºC, respectively. The samples were eluted at a flow rate of 0.3 ml min-1 using two mobile phases (A: 0.1% Formic acid in dH2O; B: 0.1% formic acid in 50:50 dH2O: Acetonitrile) with the following gradient: 0 min 5%B; 4 min 5% B; 32.00 min 100%B; 34.00 min 100% B; 36.00 min5% B; 40.00 min 5% B. For optimal electrospray ionization conditions the nebulizer pressure, drying gas temperature and drying gas were set to 45 psi, 350ºC and 3 l min-1, respectively. In addition the diode array detection (DAD) was performed at 254, 280 and 520 nm. Morin levels (internal standard) were integrated in Agilent Mass Hunter Quan software (v. B.06.00) and all the samples with deviations larger than 10% relative to the dataset mean were reinjected. For all samples, three aliquots were analysed across three different analytical batches.

Component detection, peak alignment and integration: All chromatograms were processed identically using the Agilent Software Profinder v B.06.00 (Agilent Technologies, Cheadle, Cheshire, UK) which combines peak finding and integration algorithms for high-throughput data generation. For positive and negative mode data, the batch recursive molecular feature was used with peak extraction restricted to 2.1-38.00 mins of the chromatography and a peak threshold set at 15000 counts. The algorithm was set-up to include potential adducts of +H, +Na+, +K+ and +NH4+ in positive mode and - H and +Cl- in negative mode restricting the compounds to a maximum of one charge state. The compound ion count threshold was set at two or more ions, and for alignment purposes the RT window was set at 0.70% + 0.60 mins and the mass window was set at 25 ppm + 2 mDa. A post- processing filter to restrict analysis to compounds with more than 15000 counts and present in at least 3 of the files in at least one sample group (species/line). The find-by-ion options were set to limit the extracted ion chromatogram (EIC) to the expected retention time +/- 0.40 minutes. The “Agile” algorithm was used for the integration of EIC, with a gaussian smoothing of 9 points applied before the integration with a Gaussian width of 3 points. Additionally, peak filters were set at over 15000 counts and the chromatogram formats were set to centroid when available and otherwise profile. Spectrum was extracted at 10% of peak height and excluded if the spectra within the m/z range used was above 20% of saturation. Finally a post-processing filter was applied and compounds with less than 15000 counts or present in less 3 files in at least one sample group (species/line) were excluded.

Statistical analyses: Statistical analyses of all data were performed using GenStat for Windows, 16th Edition (VSN international Ltd., Hemel Hempstead, UK). A principal component analysis (PCA), based on the correlation matrix was applied to all the QC samples to ensure that the blank, reference samples, and berry samples were well separated (data not shown). Two separate analysis of the metabolomics dataset on the berry samples were carried out for the positive and negative dataset. A PCA, using the correlation matrix, was used to generate PCA plots of the first 4 principal components, which were subsequently used for selecting the species samples providing the most separation between samples.

Yeast plasmids, strains and transformation: p426_GAL1pr-GFP-Aβ42 was generated by inserting the sequence GFP-Aβ42, obtained by the double digestion of p416_GPDpr-GFP-Aβ42 (kindly provided by Ian Macraedie, RMIT University, AU) with BamHI/SmaI, into the p426 vector. p425_GAL1pr- GFP-Aβ42 was generated by subcloning the sequence GAL1pr-GFP-Aβ42 into the SacI/HindIII restriction sites of p425 vector. S. cerevisiae strains used in this study are BY4742 erg6 MATa his3Δ1 leu2Δ0 lys2Δ0 ura3Δ YBR082c::kanMX4 (EUROSCARF) and BY4742_CDRE-lacZ MATa his3 leu2 lys2 ura3 aur1::AUR1-C-4xCDRE-lacZ (Prescott et al, 2010).

Yeast growth conditions and extract treatments: Synthetic complete (SC) medium [0.67% (w/v) yeast nitrogen base without amino acids (YNB) (Difco) and 0.79 g l-1 complete supplement mixture (CSM) (QBiogene)], containing 2% (w/v) glucose was used for growth of Crz1-reporter strain. Crz1

activation was induced with 1.8 mM MnCl2 (Stathopoulos & Cyert). FK506 (Cayman Chemicals) was used as positive control. Synthetic dropout (SD)-LEU-URA media [0.67% (w/v) YNB and 0.54 g -1 l 6-amino acid dropout CSM-ADE-HIS-LEU-LYS-TRP-URA (QBiogene)], supplemented with standard concentrations of the required amino acids and containing 1% (w/v) raffinose, were used for growth of the AD yeast model. The repression or induction of disease protein expression was carried out in medium containing glucose (control, disease-protein OFF) and galactose (disease-protein ON) at a final concentration of 2% (w/v). A pre-inoculum was prepared in appropriated raffinose or glucose (only for Crz1-activation model) medium and cultures were incubated overnight at 30°C under orbital shaking. Cells were diluted in fresh medium and cultures were incubated under the same conditions until the optical density at 600 nm (OD600 nm) reached 0.5 ± 0.05 (log growth phase). To ensure synchronized cells cultures, cell suspensions were further diluted according to the equation ODi x Vi = (ODf/(2(t/gt)) x Vf, where ODi = initial optical density of the culture, Vi = initial volume of culture, ODf = final optical density of the culture, t = time (usually 16 h), gt = generation time of the strain and Vf = final volume of culture. Readings were performed in a 96-well microtiter plate using a Biotek Power Wave XS plate spectrophotometer.

Growth assays: Yeast cultures were diluted to OD600 nm 0.12 ± 0.012 in fresh medium supplemented or not with the indicated concentrations of extracts in a 96-well microtiter plate. After 2 h incubation

at 30ºC, cultures were further diluted to OD600 nm 0.03 ± 0.003 in medium containing glucose or galactose and supplemented or not with extracts. The cultures were then incubated at 30°C with shaking for 24 h or 48 h (for the AD model) and cellular growth was kinetically monitored hourly by measuring OD600 nm. The areas under the curve (AUC) were integrated using the Origin software (OriginLab, Northampton, MA).

β–Galactosidase assays: For quantitative measurements of β–galactosidase activity, OD600 nm of cells cultures were recorded just before cell lysis. Cells were then incubated with Y-PER Yeast Protein Extraction Reagent (ThermoFisher Scientific) in 96-well microtiter plates for 20 min at 37ºC, after -1 which LacZ buffer containing 4 mg l ONPG was added and plates incubated at 30ºC. The OD420 nm

and OD550 nm was periodically monitored using a Biotek Power Wave XS Microplate Spectrophotometer until the development of the yellow color. Miller units were calculated as described previously (Garcia et al, 2016).

Fractionation of bioactive fruit extracts: The freeze-dried extract of selected berries (eg Rubus genevieri, and Rubus idaeus) were re-suspended in Milli-Q water, filtered and adsorbed onto a semi- preparation column packed with a C18 resin (PREP C18 55-105 μm 125Å, Waters Co. USA) using an AKTA Explorer (GE USA). 0.1% Formic Acid (Sigma, USA) in Milli-Q water was chosen as mobile phase A and Methanol (Chromasolv purity≥99.9%, Sigma, USA) as mobile phase B. The approach was modified for the different berry extracts (Rubus genevieri, Rubus idaeus) to achieve proper fractionation, by tuning the hydrophobicity of the mobile phase (water-methanol). Modifying loading and elution mobile phase composition and gradient profile served to generate a maximum number of well identifiable peak/fractions for further bio-activity testing (ranging from 12 to 28 fractions). Fractions were brought under vacuum using a rapid-vac (Labconco, USA) to remove methanol for further bioactivity testing.

References

Garcia G, Santos CN do, Menezes R (2016) High-Throughput Yeast-Based Reporter Assay to Identify Compounds with Anti-inflammatory Potential. In: Matthiesen R (ed) Proteostasis. Springer, New York, USA, pp 441–452

Prescott TAK, Ariño J, Kite GC, Simmonds MSJ (2012) Inhibition of human calcineurin and yeast calcineurin-dependent gene expression by Jasminum humile leaf and root extracts. J Ethnopharmacol 140:293–297. doi: 10.1016/j.jep.2012.01.020